HIST1H4A (Ab-3) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

Target Protein: HIST1H4A (UniProt: P62805), a replication-dependent histone involved in DNA compaction and transcriptional regulation .

PropertyDetails
ClonalityPolyclonal or monoclonal (depending on product variant) .
Host SpeciesRabbit (most common) .
ReactivityHuman, Mouse, Rat, Cow, S. cerevisiae .
SpecificityDetects post-translational modifications (PTMs) like acetylation (K5, K12, K16) or methylation (K20) .
ApplicationsWestern blot (WB), Immunohistochemistry (IHC), ChIP, Immunofluorescence (IF), ELISA .

Chromatin Studies

  • Epigenetic Modifications: Used to map acetylation (e.g., K5, K12, K16) and methylation marks on histone H4, which regulate gene expression .

    • Example: Detection of H4K5 acetylation in HeLa cells treated with sodium butyrate .

  • ChIP Assays: Validated for chromatin immunoprecipitation to study histone-DNA interactions .

Disease Research

  • Autoimmunity: HIST1H4A-IgG autoantibodies are diagnostic markers for systemic lupus erythematosus (SLE), showing 95% sensitivity and 90% specificity .

  • Cancer: Detects histone H4 in FFPE sections of breast and colon carcinomas .

Functional Insights

  • Antimicrobial Role: Histone H4 exhibits antimicrobial activity against S. aureus and P. acnes, confirmed via neutralizing antibody assays .

  • Cross-Reactivity Challenges: Some anti-H4 antibodies show off-target binding to similar PTMs (e.g., H3K27me3) , underscoring the need for rigorous validation.

Diagnostic Performance

  • SLE Diagnosis: In a cohort of 153 SLE patients, HIST1H4A-IgG improved diagnostic accuracy from 86% (standard antibodies) to 89% when combined with SA parameters .

Western Blot Analysis

Sample TypeResult
HeLa Cell LysateDetects a 12 kDa band corresponding to acetylated H4 (K5) .
Calf Thymus ExtractSpecific binding to H4; competition assays with acetyl-K16 peptide block signal .

Immunohistochemistry

  • Nuclear staining in human breast carcinoma and normal scalp sebocytes .

Critical Considerations

  • PTM-Specificity: Antibodies like ITK0013 (acetyl-K12) or ab7311 (pan-H4) require validation using peptide blocking or knockout controls .

  • Buffer Compatibility: Performance varies with immunoblot buffers (e.g., reducing conditions affect epitope recognition) .

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 order. Delivery time may vary depending on the chosen purchase method and location. For specific delivery timelines, 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 serves as a core component of the nucleosome, a fundamental structure responsible for packaging and compacting DNA into chromatin. This packaging process limits the accessibility of DNA to cellular machinery requiring DNA as a template. Consequently, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is regulated through a complex system of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Studies demonstrate 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), are involved in DNA damage repair. H3K36me3 stimulates H4K16ac following a DNA double-strand break, and this process requires the involvement of SETD2, LEDGF, and KAT5 (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5). PMID: 28546430
  3. Data indicates 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 elevated levels of N-myc transcription factor in these cells. PMID: 24481548
  9. Data suggests that histone assembly during the G1 phase is limited 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 human genome. PMID: 22894908
  11. SRP68/72 heterodimers are identified as major nuclear proteins whose binding to 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. Research indicates that global histone H3 and H4 modification patterns are potential markers for 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 enhancing the binding affinity of HIRA to H3.3-H4 and reducing association of CAF-1 with H3.1-H4. PMID: 21724829
  16. Imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are linked to global histone H4 modifications. PMID: 20949922
  17. Our findings reveal the molecular mechanisms by which DNA sequences within specific gene bodies are sufficient to nucleate the monomethylation of histone H4 lysine 200, which subsequently reduces gene expression by half. PMID: 20512922
  18. Histone H4 is downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  19. Low levels of histone acetylation are associated with the development and progression of gastric carcinomas, potentially through alteration of gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation levels of histone H4 protein are closely correlated. PMID: 15095300
  21. Peptidylarginine deiminase 4 (PAD4) regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine. Research suggests that PAD4 mediates gene expression by regulating Arg methylation and citrullination in histones. PMID: 15345777
  22. Lack of biotinylation of K12 in histone H4 represents an early signaling event in response to double-strand breaks. PMID: 16177192
  23. The incorporation of acetylated histone H4-K16 into nucleosomal arrays inhibits the formation of compact 30-nanometer-like fibers and hinders 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 promoting differentiation through a gene-specific modification of histone H4 arginine methylation and acetylation levels. PMID: 16782888
  26. There is a connection 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 the recruitment of various regulatory factors that can alter chromatin states for transcription regulation. PMID: 17548343
  28. Brd2 bromodomain 2 exists as a monomer in solution and dynamically interacts with H4-AcK12. Additional secondary elements in the long ZA loop may be a common feature of BET bromodomains. PMID: 17848202
  29. Spermatids Hypac-H4 impairment in mixed atrophy was not further deteriorated by AZFc region deletion. PMID: 18001726
  30. The interaction between SET8 and PCNA links H4-K20 methylation with DNA replication. PMID: 18319261
  31. H4K20 monomethylation and PR-SET7 are crucial for L3MBTL1 function. PMID: 18408754
  32. Elevated expression of acetylated H4 is more prevalent in aggressive compared to indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Our findings highlight a significant role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results indicate that acetylation of histone H4 K16 during the S-phase leads to early replicating chromatin domains acquiring the H4K16ac-K20me2 epigenetic label, which persists throughout mitosis and is deacetylated in the early G1-phase of the subsequent cell cycle. PMID: 19348949
  35. Acetylated H4 is overexpressed in diffuse large B-cell lymphoma and peripheral T-cell lymphoma relative to normal lymphoid tissue. PMID: 19438744
  36. The release of histone H4 through holocrine secretion from the sebaceous gland may play a crucial role in innate immunity. PMID: 19536143
  37. Histone modification, including PRC2-mediated repressive histone marker H3K27me3 and active histone marker acH4, may be involved in CD11b transcription during the reprogramming of HL-60 leukemia cells to terminal differentiation. PMID: 19578722
  38. A role for Cdk7 in regulating elongation is further supported by enhanced histone H4 acetylation and diminished histone H4 trimethylation on lysine 36—two hallmarks of elongation—within genes when the kinase is inhibited. PMID: 19667075
  39. Data revealed 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. Research directly implicates BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response. PMID: 19818714

Show More

Hide All

Database Links

HGNC: 4781

OMIM: 142750

KEGG: hsa:121504

STRING: 9606.ENSP00000367034

UniGene: Hs.143080

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

Q&A

What is HIST1H4A and why is it important in epigenetic research?

HIST1H4A (Histone Cluster 1, H4a) is a member of the histone H4 family, one of the core histones involved in nucleosome structure formation. Histones undergo various post-translational modifications (PTMs) that regulate chromatin structure and gene expression, making them critical components in epigenetic regulation. HIST1H4A antibodies are essential tools for investigating these modifications, allowing researchers to track specific histone marks across the genome and correlate them with transcriptional states and other chromatin features. This provides insights into fundamental epigenetic mechanisms controlling gene expression, cell differentiation, and disease processes .

What are the key post-translational modifications of HIST1H4A that can be detected with specific antibodies?

HIST1H4A can undergo multiple post-translational modifications, with specific antibodies available for each modification site. The most commonly studied modifications include:

  • Acetylation at Lysine 12 (acLys12) - involved in transcriptional activation

  • Acetylation at Lysine 16 (acLys16) - associated with chromatin relaxation

  • Acetylation at Lysine 8 (acLys8) - linked to active transcription

  • Methylation at Lysine 20 (meLys20) - associated with heterochromatin formation

  • Acetylation at Lysine 56 (acLys56) - implicated in DNA damage repair

These modifications can be detected using specific antibodies that recognize both the histone and the particular modification, with each offering insights into distinct biological processes .

How do I select the appropriate HIST1H4A antibody for my specific research application?

Selecting the appropriate HIST1H4A antibody requires consideration of several factors:

  • Target modification: Determine which specific modification (e.g., acLys12, meLys20) is relevant to your research question

  • Application compatibility: Verify that the antibody has been validated for your intended application (ChIP, IF, WB, ICC)

  • Host species: Consider potential cross-reactivity with secondary antibodies in your experimental system

  • Clonality: Polyclonal antibodies offer broader epitope recognition but may have batch variation; monoclonal antibodies provide consistency but may have more limited epitope recognition

  • Validation data: Review specificity testing data, especially for PTM-specific antibodies

  • Cross-reactivity: Ensure the antibody recognizes your species of interest

For chromatin immunoprecipitation studies, antibodies that have been specifically validated for ChIP applications should be selected, as not all antibodies perform equally across different techniques .

What are the optimal conditions for using HIST1H4A antibodies in Chromatin Immunoprecipitation (ChIP) experiments?

For optimal ChIP results with HIST1H4A antibodies, consider the following protocol guidelines:

  • Sample preparation: Use approximately 4 × 10^6 cells yielding 10 μg of chromatin per immunoprecipitation

  • Antibody amount: Use 10 μl of antibody per immunoprecipitation reaction

  • Crosslinking: Optimize formaldehyde crosslinking time (typically 10-15 minutes) to preserve protein-DNA interactions

  • Sonication: Adjust sonication conditions to generate DNA fragments between 200-500 bp

  • Controls: Include appropriate controls (IgG control, input sample, positive control loci)

  • Washing conditions: Use stringent washing buffers to reduce background while preserving specific interactions

  • Validation: Verify enrichment at known target sites using qPCR before proceeding to sequencing

For antibodies targeting specific modifications such as H4K12ac, it is critical to validate specificity using peptide competition or knockout controls to ensure the signal is truly representative of the targeted modification .

How should I optimize immunofluorescence protocols when using HIST1H4A antibodies?

When optimizing immunofluorescence protocols with HIST1H4A antibodies, follow these recommendations:

  • Fixation: Use 4% paraformaldehyde for 10-15 minutes at room temperature to preserve nuclear architecture

  • Permeabilization: Apply 0.5% Triton X-100 for 10 minutes to allow antibody access to nuclear proteins

  • Blocking: Block with 5% BSA or 10% normal serum from the secondary antibody host species

  • Antibody dilution: Start with recommended dilutions (1:50-1:200 for most HIST1H4A antibodies, or 1:200-1:800 for higher sensitivity antibodies)

  • Incubation: Incubate with primary antibody overnight at 4°C for optimal binding

  • Washing: Perform extensive washing steps (at least 3×5 minutes) with PBS-T

  • Antigen retrieval: Consider mild antigen retrieval methods if signal is weak

  • Counterstaining: Use DAPI for nuclear visualization and appropriate markers for colocalization studies

Titering the antibody concentration is crucial for optimal signal-to-noise ratio, and preparation of matched samples (treated vs. control) should be processed simultaneously to allow for direct comparison .

What validation steps should be performed to ensure antibody specificity for modified HIST1H4A?

Thorough validation of HIST1H4A modification-specific antibodies is critical for reliable experimental outcomes. Implement these key validation steps:

  • Peptide arrays: Test antibody reactivity against a panel of modified and unmodified histone peptides to confirm specificity for the target modification

  • Peptide competition assay: Pre-incubate antibody with excess target peptide to confirm signal extinction

  • Western blot analysis: Verify single band recognition at the expected molecular weight (~11-14 kDa for histone H4)

  • Dot blot titration: Test antibody against decreasing amounts of modified peptides to determine sensitivity

  • Immunoprecipitation-mass spectrometry: Confirm antibody pulls down the intended histone with the specific modification

  • Knockout/knockdown validation: Compare signal between wild-type and cells lacking the target modification

  • Internally calibrated ChIP (ICeChIP): Use spike-in standards with known modification states to quantitatively assess antibody specificity

Research has shown that many commercially available histone modification antibodies exhibit cross-reactivity with other modifications or fail to discriminate between similar modifications (e.g., different methylation states), highlighting the importance of rigorous validation .

How can I address high background issues when using HIST1H4A antibodies in immunofluorescence or ChIP experiments?

High background is a common challenge when working with histone antibodies. Implement these strategies to reduce background:

For Immunofluorescence:

  • Increase blocking time/concentration: Use 5-10% BSA or normal serum for 1-2 hours

  • Optimize antibody concentration: Perform a dilution series to find optimal concentration (1:200-1:800)

  • Extend washing steps: Increase number and duration of washes with PBS-T

  • Use highly purified antibodies: Antigen-affinity purified antibodies typically give cleaner results

  • Add protein A/G pre-clearing step: Remove non-specific binding components

  • Test alternative fixation methods: Some epitopes are sensitive to overfixation

For ChIP:

  • Increase stringency of wash buffers: Adjust salt concentration in wash buffers

  • Pre-clear chromatin: Incubate with protein A/G beads before adding antibody

  • Block beads: Pre-block beads with BSA and non-specific DNA

  • Validate antibody specificity: Ensure the antibody doesn't cross-react with similar modifications

  • Include appropriate controls: IgG controls help identify non-specific binding

High background may also indicate cross-reactivity with similar modifications, particularly for antibodies targeting specific lysine residues with similar surrounding sequences .

What are the common pitfalls in data interpretation when using HIST1H4A modification-specific antibodies?

When interpreting data from experiments using HIST1H4A modification-specific antibodies, be aware of these common pitfalls:

  • Antibody cross-reactivity: Many antibodies show reactivity to similar modifications, leading to misattribution of signals. For example, antibodies targeting different methylation states (me1, me2, me3) often cross-react, creating false patterns of distribution

  • Context-dependent epitope accessibility: Surrounding modifications may block antibody access to the target site, creating false negatives in regions with combinatorial modifications

  • Quantification limitations: Standard ChIP-seq provides relative rather than absolute quantification of modifications, making comparisons between different modifications challenging

  • Normalization issues: Improper normalization to input or control samples can create artificial patterns of enrichment

  • Biological misinterpretation: The presence of a modification does not always correlate with its expected function (e.g., H3K4me3 typically marks active promoters but can occur at inactive genes)

  • Technical artifacts: Batch effects, sample preparation differences, or sequencing biases can be misinterpreted as biological signal

The development of internally calibrated ChIP (ICeChIP) has revealed that many antibodies widely used for histone methylation studies show poor specificity for their intended targets, potentially invalidating some established paradigms in the literature .

How can I quantitatively assess the abundance of HIST1H4A modifications across different experimental conditions?

To quantitatively assess HIST1H4A modifications across different conditions, consider these methodological approaches:

  • Internally Calibrated ChIP (ICeChIP): Incorporate nucleosomes with known modifications as spike-in controls to enable absolute quantification of modification abundance. This technique allows for direct comparison between samples and accurate measurement of global PTM abundance changes

  • ChIP-Rx: Use spike-in chromatin from a different species as an internal reference to normalize between samples

  • Mass Spectrometry:

    • Use targeted MS approaches with isotopically labeled peptide standards

    • Apply Multiple Reaction Monitoring (MRM) for specific modification quantification

    • Implement SILAC labeling for comparison between experimental conditions

  • Western Blot Quantification:

    • Include recombinant standards at known concentrations

    • Use fluorescent secondary antibodies for more accurate quantification

    • Apply total histone normalization

  • Quantitative Imaging Analysis:

    • Use automated image analysis software to quantify immunofluorescence signal intensities

    • Normalize to total histone or DNA content

    • Include calibration standards in each experiment

Research has shown that H3K4 methylation states (H3K4me1/2/3) exist at different global abundances (~5–20% for H3K4me1, ~1-4% for H3K4me2), highlighting the importance of quantitative approaches to distinguish biological changes from technical variation .

How can HIST1H4A antibodies be used to study the relationship between histone modifications and transcriptional regulation?

HIST1H4A antibodies enable sophisticated investigations into the relationship between histone modifications and transcriptional control through these advanced approaches:

  • Sequential ChIP (Re-ChIP): Use two successive immunoprecipitations to identify genomic regions containing co-occurring modifications (e.g., H4K12ac and H4K16ac), revealing combinatorial epigenetic codes

  • ChIP-seq with transcriptome integration: Combine HIST1H4A ChIP-seq data with RNA-seq to correlate specific modifications with transcriptional output. This approach has revealed quantitative relationships between enhancer H3K4 methylation states and promoter activity

  • TIME-ChIP (Targeted Isolation of Modified Enhancers): Use HIST1H4A antibodies to isolate specific regulatory elements and identify associated proteins or RNAs

  • CUT&RUN or CUT&Tag: Apply these higher resolution techniques with HIST1H4A antibodies to map modifications with reduced background and lower cell numbers

  • Single-cell approaches: Combine HIST1H4A antibodies with single-cell technologies to reveal cell-to-cell variation in histone modification patterns

  • Dynamic studies: Use HIST1H4A antibodies in time-course experiments following stimulus to track modification changes during transcriptional responses

Research using these approaches has demonstrated that H3K4me3 is both phenomenologically and biochemically associated with active promoters, where it is flanked by lower H3K4 methylation states, creating a characteristic pattern at transcriptionally active genes .

What insights can be gained by studying the interplay between different HIST1H4A modifications in chromatin regulation?

Studying the interplay between different HIST1H4A modifications provides critical insights into chromatin regulation mechanisms:

  • Modification crosstalk: Certain HIST1H4A modifications influence the deposition or removal of others, creating hierarchical regulatory systems. For example, acetylation at one lysine residue may influence methylation at another, establishing sequential modification patterns

  • Reader protein recruitment: Different combinations of modifications create binding platforms for specific chromatin reader proteins, affecting downstream functions. The pattern of modifications rather than individual marks determines which regulatory complexes are recruited

  • Modification dynamics: The temporal sequence of modification appearance/disappearance during cellular processes reveals causal relationships in chromatin regulation

  • Domain-specific patterns: Comparative analysis of modifications across different chromatin domains (enhancers, promoters, gene bodies) illuminates domain-specific regulatory mechanisms

  • Developmental transitions: Tracking modification changes during development reveals epigenetic mechanisms underlying cell fate decisions

Analysis of H3K4 methylation states has demonstrated that these modifications exist in specific patterns at regulatory elements, with H3K4me1 marking enhancers, H3K4me2 associated with tissue-specific transcription factor binding sites, and H3K4me3 enriched at active promoters, suggesting coordinated deposition mechanisms .

How can high-specificity HIST1H4A antibodies improve our understanding of enhancer-promoter interactions?

High-specificity HIST1H4A antibodies enable refined investigation of enhancer-promoter interactions through these advanced approaches:

  • Quantitative enhancer state assessment: ICeChIP with high-specificity antibodies allows precise measurement of histone modification levels at enhancers, enabling quantitative correlation with promoter activity. This approach has revealed that many widely-used antibodies yield dramatically different biological interpretations of enhancer activity

  • Enhancer classification: Accurate discrimination between active, primed, and poised enhancers requires highly specific antibodies to different HIST1H4A modification states

  • Combinatorial modification mapping: High-specificity antibodies allow accurate identification of enhancers bearing multiple modifications, revealing functional enhancer subclasses

  • Differential enhancer analysis: Comparing enhancer modification patterns across cell types or conditions requires antibodies that truly distinguish between related modifications

  • Long-range interaction studies: Combining high-specificity ChIP with chromosome conformation capture techniques (HiChIP, PLAC-seq) provides more accurate mapping of functional enhancer-promoter interactions

What are the cutting-edge applications combining HIST1H4A antibodies with other technologies?

Several innovative approaches combine HIST1H4A antibodies with emerging technologies to yield powerful new insights:

  • CUT&Tag and CUT&RUN: These techniques use antibody-directed nuclease activity to map histone modifications with higher resolution and lower background than traditional ChIP, requiring fewer cells and less sequencing depth

  • Proximity ligation approaches: Combining HIST1H4A antibodies with proximity ligation enables detection of co-occurring modifications or protein-modification interactions at specific genomic loci

  • Live-cell imaging: Modified antibody fragments (Fabs) against HIST1H4A can track dynamic changes in histone modifications in living cells

  • Mass cytometry (CyTOF): Metal-conjugated HIST1H4A antibodies allow simultaneous detection of multiple histone modifications alongside other cellular proteins at single-cell resolution

  • Spatial genomics: Integration of HIST1H4A antibodies with spatial transcriptomics or imaging approaches maps histone modification patterns within tissue architecture

  • Combinatorial indexing: High-throughput single-cell ChIP approaches using HIST1H4A antibodies reveal cell-to-cell epigenetic heterogeneity

  • CRISPR screening with epigenetic readouts: Combining CRISPR perturbations with HIST1H4A ChIP enables functional dissection of modification regulatory networks

These approaches are transforming our understanding of histone modifications from static marks to dynamic regulatory systems that respond to and direct cellular processes in context-dependent ways .

What are the optimal storage and handling conditions for maintaining HIST1H4A antibody performance?

Proper storage and handling of HIST1H4A antibodies is critical for maintaining their specificity and sensitivity:

  • Storage temperature: Store antibodies at -20°C for long-term storage and at 4°C for antibodies in frequent use (typically stable for up to 1 month at 4°C)

  • Aliquoting: Divide stock solutions into small working aliquots to avoid repeated freeze-thaw cycles, which can significantly reduce antibody activity

  • Buffer composition: Most HIST1H4A antibodies are supplied in buffers containing preservatives like 0.03% Proclin 300 to maintain stability

  • Avoiding contamination: Use sterile technique when handling antibody solutions to prevent microbial growth

  • Centrifugation before use: Briefly centrifuge antibody vials before opening to collect liquid at the bottom and avoid loss

  • Transport conditions: When transporting between laboratories, maintain cold chain using dry ice or freezer packs

  • Expiration tracking: Document receipt date and track usage to ensure antibodies are used within their effective lifetime

  • Validation frequency: Periodically re-validate antibodies, particularly polyclonal lots, as performance may change over time

Antibody performance should be validated after extended storage periods, especially for critical experiments, as some epitopes may be more susceptible to degradation than others .

How does the choice between monoclonal and polyclonal HIST1H4A antibodies impact experimental outcomes?

The choice between monoclonal and polyclonal HIST1H4A antibodies significantly impacts experimental outcomes:

CharacteristicMonoclonal AntibodiesPolyclonal Antibodies
SpecificityRecognize single epitopeRecognize multiple epitopes
Batch consistencyHigh lot-to-lot reproducibilityVariation between lots
BackgroundGenerally lower backgroundMay have higher background
SensitivityMay have lower sensitivityOften higher sensitivity due to multiple binding sites
Epitope occlusionVulnerable to epitope maskingMore tolerant of partial epitope occlusion
ProductionRecombinant production ensures continuous supplyLimited by animal immunization
ApplicationsMore consistent in quantitative applicationsBetter for detection in various applications
CostGenerally higher costOften more cost-effective

For ChIP experiments, monoclonal antibodies often provide more consistent results across experiments but may be more sensitive to fixation conditions. Polyclonal antibodies typically provide higher signal but may introduce more variability .

What considerations should be made when designing ChIP-seq experiments with HIST1H4A modification-specific antibodies?

When designing ChIP-seq experiments with HIST1H4A modification-specific antibodies, consider these critical factors:

  • Antibody validation: Verify antibody specificity using peptide arrays, competition assays, or ICeChIP approaches before proceeding to genome-wide experiments

  • Experimental controls:

    • Include input controls (pre-immunoprecipitation chromatin)

    • Use IgG negative controls

    • Consider spike-in controls for quantitative normalization

    • Include positive control loci known to contain the modification of interest

  • Sequencing depth: Target 20-30 million uniquely mapped reads for point-source modifications and 40-50 million for broadly distributed modifications

  • Replicate design: Perform at least 2-3 biological replicates to enable statistical analysis

  • Chromatin preparation:

    • Optimize crosslinking conditions for histone modifications (typically 10-15 minutes)

    • Ensure proper sonication to 200-500bp fragments

    • Use appropriate amounts of chromatin (10μg per IP) and antibody (10μl)

  • Bioinformatic analysis:

    • Apply appropriate peak calling algorithms (e.g., MACS2 for sharp peaks, SICER for broad domains)

    • Consider bin-based approaches for histone modifications with diffuse patterns

    • Account for mappability and chromatin accessibility biases

  • Data integration:

    • Plan for integration with other datasets (RNA-seq, ATAC-seq, etc.)

    • Consider performing multiple histone modification ChIPs in parallel

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.