Acetyl-HIST1H4A (K8) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

AntibodyHostClonalityApplicationsSpecies ReactivityCitations
ab45166 (Abcam)RabbitMonoclonalChIP, WB, ICC/IF, Flow Cytometry, IHC-PHuman, Mouse, Rat25+
ab15823 (Abcam)RabbitPolyclonalIHC-P, ICC/IF, ChIP, WBHuman, Cow72+
CAC15192 (Biomatik)RabbitPolyclonalELISA, ICC, IF, ChIPHumanN/A
A7258 (Abclonal)RabbitPolyclonalWB, IHC-P, IF/ICC, ChIP-seq, ELISAHuman, Mouse, Rat10+
M14495-7 (Boster Bio)RabbitMonoclonalChIP, WB, ELISA, Luminex, ICCHuman, VertebratesN/A
07-328 (Merck)RabbitPolyclonalIP, WB, ChIP-seq, DBHuman, YeastMultiple

Specificity

  • ab15823: Peptide-blocking assays confirmed exclusive recognition of acetyl-K8 in histone H4, with no cross-reactivity to acetyl-K5 (H4) or acetyl-K9 (H2A) .

  • M14495-7: No cross-reactivity observed with unmodified K8 or other acetylated lysines (K5, K12, K16, K20, K31, K91) in histone H4 .

  • A7258: Detected a 11 kDa band in Western blots of TSA-treated NIH/3T3 and C6 cells, aligning with histone H4's molecular weight .

Sensitivity

  • ab45166: Showed enhanced signal in Trichostatin A-treated HeLa cells via flow cytometry (1:20 dilution) and Western blot (1:5,000 dilution) .

  • 07-328: Validated in ChIP-seq studies for poised chromatin analysis in germline development .

Chromatin Immunoprecipitation (ChIP)

  • ab45166: Demonstrated efficacy in ChIP-seq for mapping acetylated histone H4 binding sites .

  • A7258: Recommended for ChIP with 5 μg antibody per 5–10 μg chromatin .

Western Blotting (WB)

  • ab15823: Detected a 13 kDa band in calf thymus histone lysates, consistent with acetyl-K8 modification .

  • M14495-7: Identified acetyl-K8 in sodium butyrate-treated HeLa cells at 0.5 μg/mL .

Immunohistochemistry (IHC)

  • A7258: Stained human, mouse, and rat kidney sections at 1:20 dilution with citrate buffer antigen retrieval .

  • ab15823: Produced clear nuclear staining in human breast adenocarcinoma tissues using a Leica BOND system .

Comparative Analysis of Performance

Parameterab45166ab15823M14495-7
ClonalityMonoclonalPolyclonalMonoclonal
Signal IntensityHigh (ChIP-seq)Moderate (IHC)High (WB)
Species RangeBroadLimitedVertebrates
Cross-ReactivityNone reportedNone reportedNone reported

Research Findings

  • Neurodegeneration: Ethanol-induced neonatal neurodegeneration correlated with increased H4K8 acetylation, detected using 07-328 in mouse models .

  • Cancer: RM201 (M14495-7) revealed elevated H4K8ac in sodium butyrate-treated HeLa cells, implicating acetylation in chromatin remodeling .

  • Chromatin Dynamics: ab45166 facilitated the discovery of bivalent chromatin domains during mitosis-to-meiosis transitions .

Usage Recommendations

  • Blocking Buffers: 5% NFDM/TBST recommended for ab45166 and ab15823 .

  • Antigen Retrieval: EDTA buffer (pH 9.0) for IHC with ab45166; citrate buffer (pH 6.0) for A7258 .

  • Secondary Antibodies: HRP-conjugated goat anti-rabbit IgG (1:20,000) for WB; Alexa Fluor®-488 for IF .

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 products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery time estimates.
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 function in wrapping and compacting DNA into chromatin, thereby restricting accessibility of DNA to cellular machinery that requires DNA as a template. As such, histones play a pivotal role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Studies have demonstrated that PP32 and SET/TAF-Ibeta proteins inhibit HAT1-mediated H4 acetylation. PMID: 28977641
  2. Research suggests that post-translational modifications of histones, including trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), are involved in DNA damage repair. Specifically, H3K36me3 stimulates H4K16ac upon DNA double-strand breaks. SETD2, LEDGF, and KAT5 are essential for these epigenetic changes. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  3. Data indicate that Omomyc protein co-localizes with proto-oncogene protein c-myc (c-Myc), protein arginine methyltransferase 5 (PRMT5), and histone H4 H4R3me2s-enriched chromatin domains. PMID: 26563484
  4. H4K12ac is regulated by estrogen receptor-alpha and is associated with BRD4 function and inducible transcription. PMID: 25788266
  5. Systemic lupus erythematosus appears to be linked to an imbalance in histone acetyltransferases and histone deacetylase enzymes, favoring pathological H4 acetylation. PMID: 25611806
  6. Sumoylated human histone H4 prevents chromatin compaction by inhibiting long-range internucleosomal interactions. PMID: 25294883
  7. Acetylation at lysine 5 of histone H4 is associated with lytic gene promoters during reactivation of Kaposi's sarcoma-associated herpesvirus. PMID: 25283865
  8. An increase in histone H4 acetylation caused by hypoxia in human neuroblastoma cell lines corresponds to increased levels of N-myc transcription factor in these cells. PMID: 24481548
  9. Data indicate that G1-phase histone assembly is restricted to CENP-A and H4. PMID: 23363600
  10. This study 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 act as major nuclear proteins whose binding of the histone H4 tail is inhibited by H4R3 methylation. PMID: 23048028
  12. TNF-alpha inhibition of AQP5 expression in human salivary gland acinar cells is attributed to an epigenetic mechanism involving suppression of acetylation of histone H4. PMID: 21973049
  13. Our findings suggest that global histone H3 and H4 modification patterns may serve as potential markers of tumor recurrence and disease-free survival in non-small cell lung cancer. PMID: 22360506
  14. HAT1 differentially impacts nucleosome assembly of H3.1-H4 and H3.3-H4. PMID: 22228774
  15. Phosphorylation of histone H4 Ser 47, catalyzed by the PAK2 kinase, promotes nucleosome assembly of H3.3-H4 and inhibits nucleosome assembly of H3.1-H4 by increasing the binding affinity of HIRA to H3.3-H4 and reducing association of CAF-1 with H3.1-H4. PMID: 21724829
  16. The imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4. PMID: 20949922
  17. Our findings reveal the molecular mechanisms whereby the DNA sequences within specific gene bodies are sufficient to nucleate the monomethylation of histone H4 lysine 200, which in turn reduces gene expression by half. PMID: 20512922
  18. Expression is downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  19. Low levels of histone acetylation are associated with the development and progression of gastric carcinomas, possibly through alteration of gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation levels of histone H4 protein are closely related. PMID: 15095300
  21. Peptidylarginine deiminase 4 regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine. Data suggest that PAD4 mediates gene expression by regulating Arg methylation and citrullination in histones. PMID: 15345777
  22. 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. A relationship exists 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 play novel roles in mediating recruitment of multiple regulatory factors that can alter 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 crucial for L3MBTL1 function. PMID: 18408754
  32. High expression of acetylated H4 is more prevalent in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Our findings indicate a significant role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results suggest 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
  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 critical 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 supported 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 (K8) Antibody and what specific epitope does it recognize?

Acetyl-HIST1H4A (K8) antibody specifically recognizes histone H4 when acetylated at lysine 8 (K8ac). Histone H4 is a core component of nucleosomes that wrap and compact DNA into chromatin, fundamentally regulating DNA accessibility to cellular machinery. This antibody binds specifically to the acetylated form of lysine 8 on histone H4 and does not cross-react with other acetylated lysine residues on histone H4 (such as K5, K12, K16, K20, K31, or K91) .

The specificity of this antibody is critical in epigenetic research as H4K8 acetylation represents a specific post-translational modification associated with transcriptionally active chromatin regions. Proper controls should be employed to verify specificity, including peptide competition assays with acetylated and non-acetylated peptides as demonstrated in validation studies .

What applications are validated for Acetyl-HIST1H4A (K8) antibodies and what are their performance characteristics?

Acetyl-HIST1H4A (K8) antibodies have been validated for multiple applications across different research platforms:

ApplicationWorking DilutionPerformance Notes
ChIP1μg-5μg per assayValidated for chromatin binding studies
WB0.5-2 μg/mLDetects ~11 kDa band in histone extracts
ELISA0.2-1 μg/mLSuitable for quantitative detection
ICC/IF0.5-2 μg/mL (1:100-1:1000)Nuclear localization signal
IHC-P1:50-1:200Works with formalin-fixed tissues
ChIP-seqVaries by protocolFor genome-wide acetylation profiling
Luminex0.05-1 μg/mLFor multiplex detection systems

The antibody performs best with appropriate positive controls such as histone extracts from sodium butyrate-treated cells (HDAC inhibitor that increases acetylation) and negative controls including untreated cell extracts or blocking with specific peptides .

What species reactivity has been confirmed for commercial Acetyl-HIST1H4A (K8) antibodies?

Based on the provided information, commercial Acetyl-HIST1H4A (K8) antibodies demonstrate species reactivity as follows:

Antibody SourceConfirmed ReactivityNotes
Boster (RM201)Human, VertebratesMonoclonal, rabbit host
CusabioHumanPolyclonal, rabbit host
Abcam (ab15823)Human, CowPolyclonal, ChIP-grade
ABClonal (A7258)Human, Mouse, Rat, OtherWide range predicted
Abcam (EPR16606)Human, Mouse, RatRecombinant monoclonal that recognizes multiple H4 acetylation sites (K5, K8, K12, K16)

The high conservation of histone H4 sequence across species facilitates cross-reactivity, though researchers should validate antibody performance in their specific experimental model. For evolutionarily distant organisms, epitope sequence verification is recommended prior to use .

How should ChIP experiments be designed and optimized using Acetyl-HIST1H4A (K8) antibodies?

ChIP experiments using Acetyl-HIST1H4A (K8) antibodies require careful optimization for accurate results:

Protocol Optimization:

  • Chromatin Preparation: Cross-link cells with formaldehyde (typically 1-1.5%, 10 minutes at room temperature)

  • Sonication: Fragment chromatin to 200-500 bp lengths (optimize cycles for your sonicator)

  • Antibody Amount: Use 1-5 μg per ChIP reaction as a starting point

  • Chromatin Amount: 25 μg of chromatin is a typical starting point

  • Protein A/G Beads: Use 20-30 μl of properly blocked beads

Essential Controls:

  • Input control (non-immunoprecipitated chromatin)

  • No antibody control (beads only)

  • IgG control (matched isotype)

  • Positive control regions (known H4K8ac-enriched genes)

  • Negative control regions (heterochromatic or inactive regions)

Quantification Method:

  • qPCR with primers for known regulatory regions

  • For ChIP-seq, include spike-in controls for normalization and use appropriate bioinformatic pipelines

For optimal ChIP results, cell treatment with HDAC inhibitors like sodium butyrate can serve as a positive control by increasing global H4K8 acetylation levels . Antibody specificity should be validated using peptide competition assays to ensure binding is specific to H4K8ac and not other acetylated lysines on H4 .

What sample preparation protocols are critical for optimal Western blot detection of H4K8 acetylation?

Successful Western blot detection of H4K8 acetylation requires specific sample preparation protocols:

Histone Extraction Protocol:

  • Acid Extraction: Use 0.2N HCl or specialized histone extraction kits

  • Nuclei Isolation: Alternative approach for enriching histones

  • Protein Quantification: Bradford or BCA assay adjusted for histones

  • Sample Handling: Maintain samples at 4°C and include protease and deacetylase inhibitors

Loading and Transfer Considerations:

  • Load 2.5-5 μg of histone extract per lane

  • Use 15-18% polyacrylamide gels for optimal separation of low molecular weight histones

  • Add 5-10 mM sodium butyrate to all buffers to prevent deacetylation during sample processing

  • Use PVDF membrane for transfer (0.2 μm pore size recommended)

  • Short transfer times (60-90 min) at lower voltage improve retention of small histone proteins

Detection Conditions:

  • Primary antibody dilution: 0.5-2 μg/mL

  • Blocking: 5% BSA or milk (though some researchers report BSA is superior for phospho-epitopes)

  • Include both positive control (sodium butyrate-treated cells) and negative controls (untreated extracts)

  • Expected molecular weight: approximately 11 kDa

For specificity verification, competitive blocking experiments with acetylated K8 peptides can be performed . The data shows that the H4K8ac antibody specifically recognizes sodium butyrate-treated samples with increased acetylation and this recognition can be blocked by the H4K8ac peptide but not by other acetylated histone peptides .

How can immunocytochemistry experiments be optimized for Acetyl-HIST1H4A (K8) detection?

For optimal immunocytochemistry detection of H4K8 acetylation:

Cell Preparation:

  • Culture cells on appropriate coverslips or chamber slides

  • Fixation: 4% paraformaldehyde (10 min) or methanol:acetone (1:1)

  • Permeabilization: 0.1-0.5% Triton X-100 in PBS (5-10 min)

  • Blocking: 1-5% BSA or normal serum (1 hour)

Antibody Incubation:

  • Primary antibody dilution: 1:100-1:1000 (0.5-2 μg/mL)

  • Incubation time: 1-2 hours at room temperature or overnight at 4°C

  • Secondary antibody: Species-appropriate fluorophore-conjugated antibody (typically 1:500-1:2000)

Signal Enhancement and Controls:

  • For weak signals, consider signal amplification systems

  • Include positive control (sodium butyrate-treated cells)

  • Counterstain with DAPI for nuclear visualization

  • Include secondary-only control to assess background

  • Consider co-staining with other markers (e.g., actin for cell structure visualization)

Imaging Considerations:

  • Use confocal microscopy for precise nuclear localization

  • Z-stack imaging may be necessary to capture the full nuclear signal

  • Consistent exposure settings for comparative analysis

The search results show successful ICC applications with both polyclonal and monoclonal Acetyl-HIST1H4A (K8) antibodies, with reports of nuclear localization in HeLa cells and enhanced signal in sodium butyrate-treated samples .

How does H4K8 acetylation correlate with other histone modifications in gene regulation pathways?

H4K8 acetylation operates within a complex network of histone modifications that collectively regulate chromatin structure and gene expression:

Correlation with Other Acetylation Marks:
H4K8ac often co-occurs with other acetylation marks on histone H4 (K5, K12, K16) in transcriptionally active regions. These modifications work cooperatively to neutralize the positive charge of lysine residues, weakening histone-DNA interactions and promoting an open chromatin conformation . The patterns of co-occurrence can vary by cell type and genomic region.

Functional Relationships:

  • H4K8ac is generally associated with transcriptional activation

  • It may precede or follow other histone modifications in activation cascades

  • The presence of H4K8ac alongside H3K27ac often marks active enhancers

  • H4K8ac is typically depleted in regions with repressive marks like H3K9me3 or H3K27me3

Methodological Approaches for Co-occurrence Studies:

  • Sequential ChIP (Re-ChIP) to identify co-occurrence on the same nucleosomes

  • Parallel ChIP-seq experiments with antibodies against different modifications

  • Mass spectrometry analysis of modified histones to identify modification patterns

When designing experiments to study relationships between modifications, researchers should consider using antibodies that recognize specific individual modifications (like the H4K8ac-specific antibody) rather than antibodies recognizing multiple modifications (like EPR16606 which recognizes H4 acetylated at K5, K8, K12, and K16) , unless the experimental question specifically addresses the collective presence of these modifications.

What are the recommended approaches for integrating H4K8ac ChIP-seq data with other functional genomics datasets?

Integrating H4K8ac ChIP-seq data with other genomics datasets requires systematic computational approaches:

Data Integration Framework:

  • Quality Control: Assess ChIP-seq data quality (fragment size distribution, library complexity, signal-to-noise ratio)

  • Peak Calling: Use appropriate algorithms (MACS2, HOMER) with input control normalization

  • Genome Visualization: Import data tracks into genome browsers (UCSC, IGV) alongside other genomic features

  • Multi-omics Correlation:

    • RNA-seq for gene expression correlation

    • ATAC-seq/DNase-seq for chromatin accessibility

    • Other histone marks (H3K27ac, H3K4me3, etc.)

    • Transcription factor binding sites

Analytical Methods:

  • Heatmap clustering of multiple histone modifications around transcription start sites

  • Correlation analysis between H4K8ac signal and gene expression levels

  • Motif enrichment analysis within H4K8ac peaks to identify associated transcription factors

  • Pathway enrichment analysis of genes associated with H4K8ac peaks

  • Machine learning approaches to predict functional outcomes from combinatorial histone modification patterns

Validation Strategies:

  • Confirm key findings with orthogonal methods (qPCR, ICC)

  • Test functional relationships through perturbation experiments

  • Use CRISPR-based approaches to modify specific regulatory regions

When performing ChIP-seq with Acetyl-HIST1H4A (K8) antibodies, researchers should follow established guidelines for antibody validation in ChIP applications, including peptide competition assays and testing on known positive/negative genomic regions . The specificity of the antibody for K8ac over other acetylated lysines is critical for accurate interpretation of the genomic distribution data .

How can researchers distinguish between global and gene-specific changes in H4K8 acetylation in response to experimental treatments?

Distinguishing between global and gene-specific H4K8 acetylation changes requires multi-level analytical approaches:

Global H4K8ac Assessment Methods:

  • Western Blotting: Quantifies total H4K8ac levels relative to total H4

    • Use consistent loading controls (total H4 or H3)

    • Compare band intensities across treatments

  • ELISA/Luminex: Quantitative measurement of H4K8ac in histone extracts

    • Enables precise quantification with standard curves

    • Suitable for high-throughput screening

  • Mass Spectrometry: Accurate quantification of multiple histone PTMs

    • Provides relative abundance of H4K8ac vs. unmodified H4

    • Can identify combinatorial modifications on the same histone tail

Gene-Specific H4K8ac Assessment:

  • ChIP-qPCR: Targeted approach for candidate loci

    • Normalize to input and/or to unchanged reference regions

    • Include a panel of genes representing different regulation patterns

  • ChIP-seq: Genome-wide distribution of H4K8ac

    • Differential peak calling between conditions

    • Spike-in normalization for accurate quantitative comparisons

  • CUT&RUN/CUT&Tag: Higher resolution, lower background alternatives

    • May offer improved sensitivity for detecting subtle changes

Integration Strategies:

  • Correlate global changes (Western blot) with the number and intensity of ChIP-seq peaks

  • Cluster genes based on H4K8ac response patterns

  • Integrate with transcriptomic data to correlate acetylation changes with expression changes

  • Compare with other histone acetylation marks to identify modification-specific effects

When interpreting results, researchers should consider that HDAC inhibitors like sodium butyrate typically induce global H4K8 acetylation increases , while gene-specific transcription factors might recruit HATs to specific loci. Proper controls, including both technical (antibody specificity) and biological (untreated vs. treated) , are essential for accurate interpretation.

What are the common pitfalls in ChIP experiments using Acetyl-HIST1H4A (K8) antibodies and how can they be addressed?

ChIP experiments with Acetyl-HIST1H4A (K8) antibodies can encounter several challenges:

Common Pitfalls and Solutions:

  • High Background/Low Specificity

    • Issue: Non-specific antibody binding or insufficient washing

    • Solution: Increase washing stringency, optimize antibody concentration (1-5 μg recommended) , include appropriate blocking agents, and validate antibody specificity with peptide competition assays

  • Poor Enrichment

    • Issue: Inefficient antibody binding or epitope masking

    • Solution: Optimize chromatin fragmentation (200-500 bp), verify antibody performance with positive controls (sodium butyrate-treated samples) , ensure epitope accessibility through adequate crosslink reversal

  • Variable Results Between Replicates

    • Issue: Inconsistent immunoprecipitation or chromatin preparation

    • Solution: Standardize chromatin preparation protocol, maintain consistent antibody:chromatin ratios, use automated systems where possible, include spike-in controls for normalization

  • False Negative Results

    • Issue: Low H4K8ac abundance or epitope masking

    • Solution: Consider HDAC inhibitor treatment as a positive control , optimize chromatin fragmentation, test alternative fixation conditions

  • Cross-Reactivity with Other Acetylated Lysines

    • Issue: Antibody binding to similar acetylated epitopes

    • Solution: Use validated antibodies with demonstrated specificity for K8ac over other sites , perform peptide competition controls with K8ac and other acetylated peptides (K5ac, K12ac, K16ac)

Critical Quality Controls:

  • Include mock IP (no antibody) control

  • Use IgG isotype control

  • Test known positive regions (housekeeping genes) and negative regions

  • Validate key findings with orthogonal methods

The specificity tests shown in the search results demonstrate that a proper H4K8ac antibody should recognize acetylated K8 but not unmodified K8 or other acetylated lysines (K5, K12, K16, K20, K31, or K91) in H4 , which is crucial for accurate ChIP results.

How can researchers validate Acetyl-HIST1H4A (K8) antibody specificity for their experimental system?

Thorough validation of Acetyl-HIST1H4A (K8) antibody specificity is essential for reliable experimental results:

Comprehensive Validation Strategy:

  • Peptide Competition Assays

    • Preincubate antibody with acetylated K8 peptide (specific competition)

    • Preincubate with unacetylated K8 peptide (should not compete)

    • Preincubate with peptides containing other acetylated lysines (K5ac, K12ac, K16ac) to confirm specificity

    • Perform Western blot or ChIP with competed and non-competed antibody

  • Positive and Negative Controls

    • Positive: Histone extracts from HDAC inhibitor-treated cells (sodium butyrate, TSA)

    • Negative: Untreated cells or HAT inhibitor-treated cells

    • Genetic controls: Cells with mutated K8 residue (if available)

  • Cross-Platform Validation

    • Compare results across multiple techniques (WB, ChIP, ICC)

    • Verify consistent patterns of enrichment/depletion

  • Antibody Performance Metrics

    • Signal-to-noise ratio in different applications

    • Concentration-dependent response curves

    • Batch-to-batch consistency testing

    • Cross-reactivity assessment with similar epitopes

Validation Test Panel:

Validation TestExpected Result for Specific Antibody
WB with K8ac peptide competitionSignal elimination or significant reduction
WB with K5ac/K12ac/K16ac peptide competitionMinimal effect on signal
HDAC inhibitor treatment (Western blot)Increased signal intensity
Immunoprecipitation + Mass SpecEnrichment of H4K8ac peptides
ChIP-qPCR at known targetsEnrichment at active genes, depleted at inactive regions
ImmunofluorescenceNuclear staining increased in HDAC inhibitor-treated cells

The search results show examples of antibody validation through specificity tests that confirm the Acetyl-HIST1H4A (K8) antibody recognizes only H4 acetylated at K8 and not other acetylated lysines .

What strategies can address weak or inconsistent signals when detecting H4K8 acetylation?

When facing weak or inconsistent H4K8 acetylation signals, multiple optimization strategies can be employed:

Signal Enhancement Approaches:

  • Sample Preparation Optimization

    • Include deacetylase inhibitors (sodium butyrate, TSA, nicotinamide) in all buffers

    • Optimize extraction protocols for histone enrichment

    • Minimize sample processing time to prevent modification loss

    • Consider using fresh samples rather than frozen when possible

  • Detection Method Refinement

    • Western Blot: Use high-sensitivity ECL substrates, optimize transfer conditions for small proteins, consider loading more protein (2.5-5 μg)

    • ChIP: Increase chromatin amount (up to 25 μg) , optimize sonication, extend antibody incubation time

    • ICC/IF: Implement signal amplification systems, optimize fixation (test both PFA and methanol), adjust permeabilization conditions

  • Antibody Optimization

    • Test concentration range (0.5-5 μg/mL for most applications)

    • Compare different antibody clones/sources

    • Optimize incubation conditions (time, temperature, buffer composition)

    • Consider monoclonal (higher specificity) vs. polyclonal (potentially higher sensitivity)

  • Biological Signal Enhancement

    • Treat cells with HDAC inhibitors as positive controls

    • Target cell types/conditions with naturally higher H4K8ac levels

    • Synchronize cells to capture cell-cycle dependent fluctuations

Troubleshooting Inconsistent Results:

IssuePotential CausesSolutions
Variable signal between replicatesBiological variation, inconsistent sample processingStandardize protocols, increase replicate number, include internal controls
Signal in controlsAntibody cross-reactivity, high backgroundValidate antibody specificity , optimize blocking, increase washing stringency
Decreasing signal over timeAntibody degradation, modification loss during storagePrepare fresh aliquots, add protease/deacetylase inhibitors, optimize storage conditions
Detection in incorrect cellular locationFixation artifacts, antibody cross-reactivityValidate with different fixation methods, perform competition assays

The antibody dilution recommendations from the search results (0.5-2 μg/mL for Western blot, 1:100-1:1000 for ICC/IF) provide starting points, but optimization for specific experimental systems is essential for reliable detection of H4K8 acetylation.

How should researchers interpret changes in H4K8 acetylation patterns in the context of gene expression regulation?

Interpreting H4K8 acetylation changes requires nuanced analysis within the broader epigenetic context:

Interpretative Framework:

  • Genomic Location Analysis

    • Promoter-associated H4K8ac: Typically correlates with transcriptional activation

    • Enhancer-associated H4K8ac: May indicate enhancer activation, particularly when co-occurring with H3K27ac

    • Gene body H4K8ac: Can reflect active transcriptional elongation

    • Intergenic H4K8ac: Potential enhancers or regulatory elements

  • Temporal Dynamics Considerations

    • Rapid H4K8ac changes (minutes to hours): Often precede transcriptional changes

    • Stable H4K8ac patterns: May represent established epigenetic states

    • Sequential histone modification events: H4K8ac may be part of a cascade of modifications

  • Correlation with Transcription

    • Direct correlation: H4K8ac increases accompanied by increased transcription

    • Permissive state: H4K8ac present but transcription dependent on additional factors

    • Uncoupled patterns: Changes in H4K8ac without corresponding expression changes may indicate priming or other non-transcriptional functions

  • Multi-modification Context

    • Consider H4K8ac alongside other histone H4 acetylation marks (K5, K12, K16)

    • Analyze relationship with other activation marks (H3K4me3, H3K27ac)

    • Examine mutual exclusivity with repressive marks (H3K9me3, H3K27me3)

Data Integration Approaches:

  • Correlate H4K8ac ChIP-seq data with RNA-seq from matched samples

  • Perform time-course experiments to establish causality

  • Integrate with transcription factor binding data to identify recruitment mechanisms

  • Employ genome editing to modify H4K8 residues at specific loci

The functional significance of H4K8 acetylation stems from its role in nucleosome structure and DNA accessibility. As a core component of nucleosomes, histone H4 wraps and compacts DNA, with acetylation of K8 neutralizing positive charges and potentially weakening histone-DNA interactions to facilitate transcription factor binding and gene expression .

What are the established links between H4K8 acetylation and specific cellular processes or disease states?

H4K8 acetylation has been implicated in various biological processes and pathological conditions:

Physiological Processes:

  • Transcriptional Regulation

    • H4K8ac is associated with actively transcribed genes

    • Functions in the recruitment of transcriptional machinery

    • Works cooperatively with other histone acetylation marks

  • Cell Cycle Regulation

    • Fluctuations in H4K8ac levels during different cell cycle phases

    • Potential role in DNA replication and chromosome segregation

  • Development and Differentiation

    • Dynamic H4K8ac patterns during cellular differentiation

    • Tissue-specific H4K8ac profiles reflecting developmental programs

  • Stress Response

    • Rapid changes in H4K8ac in response to environmental stressors

    • Potential role in activating stress-response genes

Pathological Associations:

  • Cancer

    • Altered H4K8ac patterns in various cancer types

    • Potential diagnostic/prognostic biomarker

    • Target of epigenetic therapies (HDAC inhibitors)

  • Neurodegenerative Disorders

    • Dysregulated histone acetylation including H4K8ac in conditions like Alzheimer's and Parkinson's

    • Therapeutic potential of restoring normal acetylation patterns

  • Inflammatory Conditions

    • H4K8ac changes at inflammatory gene loci

    • Potential involvement in cytokine gene regulation

  • Metabolic Disorders

    • Links to metabolic gene regulation

    • Crosstalk with metabolic sensors and signaling pathways

Research Applications:

  • Using H4K8ac antibodies to identify disease-specific epigenetic signatures

  • Monitoring treatment responses to epigenetic therapies

  • Developing targeted approaches to modulate specific genes through H4K8 acetylation

Understanding these connections helps researchers design meaningful experiments to investigate H4K8ac in their specific biological contexts. The availability of highly specific antibodies that distinguish H4K8ac from other acetylation sites is crucial for accurately mapping these relationships.

How can researchers distinguish between cause and consequence when studying H4K8 acetylation in gene regulatory networks?

Distinguishing causality from correlation in H4K8 acetylation studies requires sophisticated experimental designs:

Causal Relationship Determination Methods:

  • Temporal Analysis

    • Time-course experiments with fine resolution sampling

    • Track H4K8ac changes relative to transcriptional changes

    • Example approach: ChIP-seq and RNA-seq at multiple timepoints after stimulus

  • Direct Manipulation Strategies

    • HAT/HDAC Modulation: Targeted recruitment of HATs/HDACs to specific loci

      • CRISPR-dCas9 fused to HATs (p300, CBP) or HDACs to modify H4K8ac at specific loci

      • Monitor consequent transcriptional changes

    • Histone Mutation: K→R or K→Q mutations to prevent or mimic acetylation

      • Requires careful experimental design in model systems

    • Domain-Specific Inhibitors: Target specific HAT/HDAC complexes that modify H4K8

  • Reader Protein Identification

    • Identify proteins that specifically bind H4K8ac

    • Disrupt these interactions to determine functional consequences

    • Techniques: CRISPR screens, protein-protein interaction studies, bromodomain inhibitors

  • Multi-omics Integration

    • Correlate H4K8ac changes with:

      • Transcription factor binding (ChIP-seq)

      • Chromatin accessibility (ATAC-seq)

      • Transcriptional output (RNA-seq)

      • Other histone modifications

    • Use causal inference statistical approaches

Experimental Design Considerations:

Experimental ApproachStrengthsLimitationsKey Controls
Time-course analysisEstablishes temporal orderCorrelation ≠ causationParallel profiling of multiple marks
CRISPR-dCas9-HAT/HDACDirect manipulationOff-target effectsdCas9 only controls, multiple guide RNAs
Histone mutantsDirect test of functionSystem-wide effectsWild-type controls, rescue experiments
Pharmacological interventionRelatively simpleLow specificityDose-response, multiple inhibitors

To confidently establish causality, researchers should employ multiple complementary approaches and carefully control for confounding factors. The availability of highly specific H4K8ac antibodies enables precise monitoring of acetylation changes following experimental manipulations, though researchers should remain cognizant of potential cross-reactivity with other acetylation sites when interpreting results.

How are new technologies enhancing the detection and functional analysis of H4K8 acetylation?

Emerging technologies are revolutionizing how researchers study H4K8 acetylation:

Advanced Detection Technologies:

  • CUT&RUN/CUT&Tag

    • Higher signal-to-noise ratio than traditional ChIP

    • Requires less starting material (thousands vs. millions of cells)

    • Compatible with H4K8ac antibodies for improved genome-wide mapping

    • Provides higher resolution of acetylation boundaries

  • Single-Cell Epigenomics

    • scCUT&Tag, scATAC-seq with targeted antibody enrichment

    • Reveals cell-to-cell variation in H4K8ac patterns

    • Can correlate with single-cell transcriptomics for direct function assessment

  • Live-Cell Imaging

    • Antibody-derived H4K8ac sensors for real-time monitoring

    • FRET-based approaches to detect dynamic changes

    • Super-resolution microscopy for subnuclear localization

  • Mass Spectrometry Advances

    • Improved sensitivity for detecting low-abundance modifications

    • Ability to identify combinatorial modifications on the same histone tail

    • Quantitative approaches for measuring absolute acetylation levels

Functional Analysis Innovations:

  • CRISPR Epigenome Editing

    • Site-specific modulation of H4K8ac using dCas9-HAT/HDAC fusions

    • Allows direct testing of functional consequences at specific loci

    • Combinatorial editing of multiple modifications simultaneously

  • Acetylation-Specific Readers and Degrons

    • Tools to selectively bind or degrade proteins based on H4K8ac status

    • Enables temporal control of acetylation-dependent processes

  • Spatial Technologies

    • Coupling H4K8ac detection with spatial transcriptomics

    • Visualizing nuclear organization of H4K8ac domains

  • Multi-modal Single-molecule Approaches

    • Direct observation of H4K8ac dynamics on individual nucleosomes

    • Real-time correlation with transcriptional machinery recruitment

These technologies will depend on highly specific antibodies like those described in the search results , with recombinant monoclonal antibodies offering advantages in reproducibility and specificity . As these methods evolve, they promise to reveal new insights into the dynamic regulation and functional consequences of H4K8 acetylation in diverse biological contexts.

What are the most promising research directions for understanding H4K8 acetylation biology?

Several promising research directions are emerging in the field of H4K8 acetylation biology:

Regulatory Mechanisms and Writer/Eraser Dynamics

  • Identification of specific HATs and HDACs controlling H4K8 acetylation

  • Elucidation of recruitment mechanisms to specific genomic loci

  • Understanding the kinetics of H4K8 acetylation/deacetylation cycles

  • Mapping the integration of signaling pathways that regulate these enzymes

Combinatorial Histone Code Integration

  • Comprehensive mapping of H4K8ac co-occurrence with other modifications

  • Understanding functional differences between isolated H4K8ac and combined patterns with other acetylation sites (K5, K12, K16)

  • Developing predictive models for transcriptional outcomes based on modification patterns

  • Identifying reader proteins that recognize specific combinations

Cell Type-Specific Functions

  • Comparative analysis of H4K8ac landscapes across diverse cell types

  • Identifying tissue-specific regulatory roles and targets

  • Understanding context-dependent functions in different cellular environments

  • Single-cell analysis of H4K8ac heterogeneity within tissues

H4K8ac in 3D Genome Organization

  • Relationship between H4K8ac and higher-order chromatin structure

  • Role in enhancer-promoter interactions and topologically associating domains

  • Contribution to phase separation and biomolecular condensate formation

  • Integration with chromosome conformation capture technologies

Non-Canonical Functions

  • Potential roles beyond transcriptional regulation

  • Investigation of H4K8ac in DNA repair processes

  • Functions in non-coding RNA regulation

  • Potential non-histone protein targets of the same acetylation machinery

Therapeutic Applications

  • Development of small molecules to specifically modulate H4K8ac

  • Targeting writer/eraser enzymes with improved specificity

  • Biomarker applications in disease diagnosis and treatment response

  • H4K8ac-based patient stratification for precision medicine approaches

The availability of highly specific antibodies that can distinguish H4K8ac from other acetylation sites will be crucial for these research directions, particularly as techniques become more sensitive and require greater epitope specificity. Recombinant monoclonal antibodies with validated specificity offer advantages for reproducible findings across different research groups .

What experimental controls and considerations are essential when developing novel assays for H4K8 acetylation?

When developing new assays for H4K8 acetylation, researchers must implement rigorous controls and considerations:

Essential Antibody Validation Controls:

  • Specificity Verification

    • Peptide competition assays with acetylated and non-acetylated peptides

    • Testing against similar modifications (K5ac, K12ac, K16ac)

    • Western blot detection of a single band at the expected molecular weight (~11 kDa)

    • Testing in HAT/HDAC inhibitor treated samples

  • Cross-Platform Validation

    • Confirm consistent results across multiple applications

    • Compare results from different antibody clones targeting the same epitope

    • Validate key findings with orthogonal non-antibody methods when possible

Assay Development Considerations:

  • Sample Preparation Standardization

    • Consistent fixation protocols for cross-linking assays

    • Standardized extraction methods for histone proteins

    • Inclusion of deacetylase inhibitors throughout processing

    • Careful consideration of buffer compositions

  • Technical Parameters

    • Signal-to-noise optimization for each assay format

    • Linear dynamic range determination

    • Limit of detection and quantification establishment

    • Reproducibility assessment through technical and biological replicates

  • Biological Controls

    • Positive controls: HDAC inhibitor (sodium butyrate, TSA) treated samples

    • Negative controls: Untreated or HAT inhibitor treated samples

    • Genetic controls: K8R mutants (where feasible)

    • Cell type controls: Known high and low H4K8ac expressing cells

Novel Assay Calibration Approach:

Assay ParameterValidation MethodAcceptance Criteria
SpecificityPeptide competition, western blot>90% signal reduction with specific peptide, <10% with non-specific peptides
SensitivitySerial dilutions of known samplesDefined lower limit of detection with CV <20%
PrecisionIntra- and inter-assay replicatesCV <15% within plates, <20% between plates
AccuracySpike-in recovery, comparison to established methods80-120% recovery, R² >0.9 correlation with reference method
RobustnessTesting across multiple operators/labsConsistent results across testing sites

For any new technological approach, comprehensive comparison with established methods is essential. The search results provide examples of validated applications for H4K8ac antibodies , which can serve as benchmarks for novel assay development and validation.

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.