HIST1H3A (Ab-18) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HIST1H3A (Ab-18) Antibody

HIST1H3A (Histone Cluster 1 H3a) is a member of the histone H3 family, which plays a pivotal role in DNA packaging and gene regulation. The HIST1H3A (Ab-18) Antibody is a polyclonal antibody generated in rabbits using a synthetic peptide corresponding to residues surrounding Lys18 of human histone H3.1 . It is widely used to study post-translational modifications (PTMs) and chromatin dynamics, particularly in human samples .

Immunogen and Host

  • Immunogen: Peptide sequence around Lys18 of human histone H3.1 .

  • Host Species: Rabbit .

  • Clonality: Polyclonal IgG .

Specificity and Cross-Reactivity

The antibody is validated for specificity using peptide arrays and immunoassays. For example, similar antibodies targeting acetylated Lys18 (e.g., ab1191) undergo rigorous testing against modified and unmodified histone peptides to ensure minimal cross-reactivity . While direct validation data for HIST1H3A (Ab-18) is not explicitly provided, its design suggests selectivity for the Lys18 region .

Applications in Research

The HIST1H3A (Ab-18) Antibody is compatible with multiple experimental workflows:

ApplicationDetails
Immunofluorescence (IF)Detects histone H3.1 in fixed cells, often colocalized with nuclear markers like DAPI .
Immunohistochemistry (IHC)Used for chromatin structure analysis in tissue sections .
ELISAQuantifies histone H3.1 levels in lysates .

Chromatin Dynamics

Studies using analogous antibodies (e.g., acetyl-K18-specific antibodies) reveal that Lys18 modifications correlate with active transcription and DNA repair . For instance, acetylation at this site is linked to open chromatin configurations in HeLa cells .

Heterogeneity in Histone Incorporation

Single-cell analyses demonstrate heterogeneity in histone H3.1 distribution, which the HIST1H3A (Ab-18) Antibody can visualize via IF . This heterogeneity reflects variable nucleosome assembly during DNA replication .

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 your order within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery information.
Synonyms
Histone H3.1 (Histone H3/a) (Histone H3/b) (Histone H3/c) (Histone H3/d) (Histone H3/f) (Histone H3/h) (Histone H3/i) (Histone H3/j) (Histone H3/k) (Histone H3/l), HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3I, HIST1H3J, H3FA, H3FL, H3FC, H3FB, H3FD, H3FI, H3FH, H3FK, H3FF, H3FJ
Target Names
Uniprot No.

Target Background

Function
Histone H3A is a core component of the nucleosome, which serves as the fundamental unit of DNA packaging in eukaryotic cells. Nucleosomes wrap and compact DNA into chromatin, thereby limiting DNA accessibility to cellular machinery that requires DNA as a template. This regulation is crucial for various cellular processes, including transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is controlled by a complex system of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer can be influenced by inducing E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Studies have shown that increased expression of H3K27me3 during a patient's clinical course can be useful in determining whether tumors are heterochronous. PMID: 29482987
  3. Research has identified JMJD5, a Jumonji C (JmjC) domain-containing protein, as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that cause a DNA damage response. PMID: 28982940
  4. Evidence suggests that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) presents an alternative proliferative marker. PMID: 29040195
  5. These findings indicate that cytokine-induced histone 3 lysine 27 trimethylation acts as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data reveals that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, the histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Research indicates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral, and deposits histone H3.3 onto these. PMID: 28981850
  9. Experiments have shown that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl) where it functions as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This study presents, for the first time, the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG) including GBM and DIPG harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3) which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data suggest that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest whereas the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Research suggests that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within the H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications caused by traffic-derived airborne particulate matter exposures in leukocytes. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. On the other hand, histone 3 mutations do not seem to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. Taken together, the authors verified that histone H3 is a real substrate for GzmA in vivo in the Raji cells treated by staurosporin. PMID: 26032366
  28. We conclude that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is important for stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) serves a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Data indicate that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data indicate that inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. The authors report that de novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays is regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132

Show More

Hide All

Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is HIST1H3A (Ab-18) Antibody and what is its target?

HIST1H3A (Ab-18) is a rabbit polyclonal antibody specifically designed to target the histone H3.1 protein, with its epitope centered around lysine 18 (K18). The antibody recognizes the peptide sequence around this site derived from Human Histone H3.1. Histone H3 is a core component of nucleosomes that wrap and compact DNA into chromatin, thereby regulating DNA accessibility to cellular machinery required for transcription, replication, and repair . This antibody is particularly valuable for studying post-translational modifications at the K18 position, which plays an important role in epigenetic regulation.

What experimental applications is the HIST1H3A (Ab-18) Antibody validated for?

The HIST1H3A (Ab-18) polyclonal antibody has been validated for multiple experimental techniques including:

  • Enzyme-Linked Immunosorbent Assay (ELISA)

  • Western Blotting (WB)

  • Immunohistochemistry (IHC)

  • Immunofluorescence (IF)

  • Immunoprecipitation (IP)

This versatility makes it suitable for diverse experimental approaches across epigenetic research. When designing experiments, researchers should consider the specific application needs and optimize antibody dilutions accordingly for each technique to ensure optimal signal-to-noise ratio.

What species reactivity has been confirmed for this antibody?

The antibody has been confirmed to react with human (Homo sapiens) and rat (Rattus norvegicus) samples . When working with other species, cross-reactivity is possible due to the high conservation of histone H3 across species, but validation experiments should be performed before proceeding with full-scale studies. For species not explicitly listed, researchers should conduct preliminary experiments with appropriate positive and negative controls to validate cross-reactivity.

How can I validate the specificity of HIST1H3A (Ab-18) Antibody for my experimental system?

Validating antibody specificity is crucial for accurate data interpretation. For HIST1H3A (Ab-18) Antibody, consider these methodological approaches:

  • Peptide Competition Assay: Pre-incubate the antibody with increasing concentrations of the immunizing peptide before application to your samples. Signal reduction confirms specificity.

  • Knockdown/Knockout Controls: Compare staining in wild-type versus H3.1-depleted samples (using siRNA, CRISPR, etc.).

  • Cross-Reactivity Assessment: Test against recombinant histone variants (H3.2, H3.3) to confirm specificity for H3.1.

  • Histone Antibody Specificity Database: Consult this resource (http://www.histoneantibodies.com) to evaluate reported cross-reactivity patterns of various histone antibodies .

  • Western Blot Molecular Weight Verification: Confirm a single band at ~17kDa corresponding to histone H3.

These validation steps are essential as histone antibodies can exhibit cross-reactivity with similar epitopes or recognize unmodified regions, potentially leading to false positive results .

How should I optimize extraction protocols for studying histone modifications with this antibody?

Effective histone extraction is critical for accurate detection of modifications. Consider this optimized protocol:

  • Cell Harvest: Collect cells at appropriate density (70-80% confluence for adherent cells).

  • Acid Extraction:

    • Lyse cells in hypotonic buffer (10mM Tris-HCl pH 8.0, 1mM KCl, 1.5mM MgCl₂, 1mM DTT) with protease inhibitors

    • Extract histones with 0.2N HCl for 30 minutes on ice

    • Centrifuge at 13,000g for 10 minutes at 4°C

    • Neutralize supernatant with 1/10 volume of 2M NaOH

  • Phosphatase Inhibitors: Include sodium fluoride (10mM) and sodium orthovanadate (1mM) to preserve phosphorylation states.

  • Deacetylase Inhibitors: Add sodium butyrate (5mM) and nicotinamide (5mM) to preserve acetylation states when studying K18 acetylation .

  • Quantification: Determine protein concentration using Bradford assay rather than BCA, as the latter can be affected by acid.

This extraction protocol maximizes yield while preserving histone modifications for subsequent analysis. For chromatin immunoprecipitation (ChIP) applications, crosslinking steps should be incorporated before extraction.

What are potential sources of false positives/negatives when using this antibody for chromatin immunoprecipitation (ChIP)?

When using HIST1H3A (Ab-18) antibody for ChIP, several factors can affect data reliability:

Sources of False Positives:

  • Cross-reactivity with similar histone modifications (e.g., antibodies against H3K27me3 can cross-react with H3K4me3-marked histones)

  • Epitope recognition in the absence of target PTM

  • Insufficient washing stringency

  • Non-specific binding to beads or reagents

Sources of False Negatives:

  • Epitope masking by neighboring modifications

  • Insufficient chromatin fragmentation

  • Suboptimal antibody concentration

  • Degradation of histone modifications during sample preparation

Methodological Solutions:

  • Include appropriate controls (IgG negative control, input normalization)

  • Validate antibody specificity with peptide arrays before ChIP experiments

  • Optimize sonication conditions to achieve fragments of 200-500bp

  • Use quantitative PCR with multiple primer sets for validation

  • Consider complementary techniques (e.g., CUT&RUN) for confirmation

Cross-validation with orthogonal methods is essential to mitigate these technical challenges and ensure reliable data interpretation .

How should I design experiments to differentiate between closely related histone variants using this antibody?

Distinguishing between histone H3 variants (H3.1, H3.2, H3.3) presents a significant challenge due to their high sequence similarity. For experiments with HIST1H3A (Ab-18) antibody:

  • Sequential Immunoprecipitation:

    • First IP with variant-specific antibody

    • Elute and perform second IP with HIST1H3A (Ab-18)

    • Compare enrichment patterns

  • Combined ChIP-Seq and RNA-Seq Analysis:

    • ChIP-Seq with HIST1H3A (Ab-18) antibody

    • RNA-Seq to determine expression of H3 variants

    • Correlate findings to identify variant-specific patterns

  • Spike-in Controls:

    • Add known quantities of recombinant H3 variants

    • Process alongside experimental samples

    • Use for quantitative normalization

  • Site-Directed Mutagenesis:

    • Express tagged H3 variants with specific mutations

    • Perform ChIP with tag-specific and HIST1H3A antibodies

    • Compare binding patterns

The most reliable approach combines multiple methodologies to overcome the inherent limitations of antibody-based discrimination between highly similar protein variants.

What normalization methods are recommended when quantifying histone modifications across different samples?

Proper normalization is essential for meaningful comparisons of histone modifications across samples. Consider these methodological approaches:

  • Total H3 Normalization:

    • Process parallel samples with HIST1H3A (Ab-18) and total H3 antibodies

    • Calculate modification level as ratio of modified H3 to total H3

    • This controls for differences in nucleosome density and extraction efficiency

  • Internal Standard Method:

    • Include stable isotope-labeled histone peptides of known concentration

    • Use as internal standards for absolute quantification

    • Particularly valuable for mass spectrometry-based analyses

  • Multiple Reference Gene Approach:

    • Normalize to multiple genomic regions with stable H3 occupancy

    • Calculate geometric mean of reference signals

    • Reduces bias from single reference regions

  • Global Normalization for Sequencing Data:

    • For ChIP-seq data, normalize to total mapped reads or spike-in controls

    • Consider quantile normalization for comparing datasets

    • Account for library composition biases

Normalization MethodAdvantagesLimitationsBest For
Total H3Controls for nucleosome densityRequires parallel samplesWestern blot, ChIP-qPCR
Internal StandardAbsolute quantificationCostly, technically demandingMass spectrometry
Multiple ReferenceReduces single reference biasRequires validated stable regionsChIP-qPCR
GlobalHandles genome-wide dataMay mask global changesChIP-seq analysis

The Histone H3 PTM Multiplex Assay offers an efficient approach for normalizing to total H3 levels across different sample preparations, enabling reliable analysis of relative changes in histone PTMs .

How can I resolve contradictory results between Western blotting and immunofluorescence using this antibody?

Discrepancies between Western blot and immunofluorescence results with HIST1H3A (Ab-18) antibody may arise from several technical factors. Here's a systematic approach to reconcile such contradictions:

  • Epitope Accessibility Analysis:

    • Western blotting denatures proteins, potentially exposing hidden epitopes

    • In immunofluorescence, native conformation may mask certain epitopes

    • Solution: Compare results with different fixation methods (paraformaldehyde, methanol, acetone)

  • Modification-Specific Differences:

    • PTMs can be cell-cycle dependent or restricted to specific nuclear compartments

    • Western blot represents population average; IF reveals single-cell heterogeneity

    • Solution: Synchronize cells and perform time-course experiments

  • Technical Validation:

    • Perform peptide competition assays in both techniques

    • Test multiple antibody dilutions and incubation conditions

    • Include positive and negative controls (e.g., cells treated with HDAC inhibitors for acetylation studies)

  • Cross-Platform Correlation:

    • Quantify signal intensity across both methods

    • Plot correlation between techniques for multiple samples

    • Identify sample-specific or technique-specific patterns

When discrepancies persist, results should be validated with a third technique (e.g., flow cytometry, mass spectrometry) to determine which approach more accurately represents the biological reality.

What are common causes of high background when using HIST1H3A (Ab-18) antibody in immunofluorescence?

High background in immunofluorescence experiments can compromise signal-to-noise ratio and data interpretation. For HIST1H3A (Ab-18) antibody, consider these methodological solutions:

Common Causes and Solutions:

  • Insufficient Blocking:

    • Extend blocking time to 2 hours at room temperature

    • Use 5% BSA or 10% normal serum from secondary antibody host species

    • Add 0.1-0.3% Triton X-100 to blocking buffer for better penetration

  • Non-specific Antibody Binding:

    • Pre-adsorb antibody with acetone powder from relevant species

    • Dilute antibody in blocking buffer containing 0.1% Tween-20

    • Optimize antibody concentration (typically 1:500-1:2000)

  • Fixation Issues:

    • Compare different fixation methods (4% PFA, methanol, or combination)

    • Avoid overfixation which can increase autofluorescence

    • Include antigen retrieval step (10mM sodium citrate, pH 6.0 at 95°C for 20 minutes)

  • Autofluorescence:

    • Treat with 0.1% Sudan Black in 70% ethanol for 20 minutes

    • Include 10mM NH₄Cl in wash buffer to quench aldehyde-induced fluorescence

    • Use appropriate filters to minimize autofluorescence detection

  • Secondary Antibody Cross-reactivity:

    • Use highly cross-adsorbed secondary antibodies

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

    • Consider using directly conjugated primary antibodies

Systematic optimization of these parameters should significantly reduce background while preserving specific nuclear staining of histone H3.

How can I improve signal detection in Western blots using HIST1H3A (Ab-18) antibody?

Optimizing Western blot protocols for histone detection presents unique challenges due to their small size (~17kDa) and various post-translational modifications. For HIST1H3A (Ab-18) antibody:

Protocol Optimization Steps:

  • Sample Preparation:

    • Use acid extraction method for histone enrichment

    • Load 10-20μg of acid-extracted histones or 30-50μg of whole cell lysate

    • Add phosphatase and deacetylase inhibitors to preserve modifications

  • Gel Electrophoresis:

    • Use 15-18% SDS-PAGE gels for better resolution of small proteins

    • Consider Triton-Acid-Urea (TAU) gels for separation of modified histones

    • Run at lower voltage (80-100V) to prevent overheating

  • Transfer Optimization:

    • Use 0.2μm PVDF membrane instead of 0.45μm

    • Transfer at 30V overnight at 4°C

    • Add 0.1% SDS to transfer buffer to improve histone transfer

  • Blocking and Antibody Incubation:

    • Block with 5% BSA in TBST (not milk, which contains phosphatases)

    • Dilute HIST1H3A (Ab-18) antibody 1:1000 in blocking buffer

    • Incubate overnight at 4°C with gentle agitation

  • Detection Enhancement:

    • Use high-sensitivity ECL substrates or fluorescent secondary antibodies

    • Consider signal amplification systems for low-abundance modifications

    • Optimize exposure times using incremental exposures

This optimized protocol typically yields clean, specific bands at ~17kDa corresponding to histone H3, with minimal background or non-specific signals.

What controls should be included when studying dynamic histone modifications with this antibody?

When investigating dynamic histone modifications using HIST1H3A (Ab-18) antibody, comprehensive controls are essential for data validation and interpretation:

Essential Experimental Controls:

  • Technical Controls:

    • Antibody specificity control (peptide competition assay)

    • Loading control (total protein stain or housekeeping protein)

    • Secondary antibody-only control (to assess non-specific binding)

  • Biological Modification Controls:

    • Positive control: Cells treated with HDAC inhibitors (e.g., TSA, sodium butyrate) to increase acetylation

    • Negative control: Cells with CRISPR/siRNA knockdown of relevant histone acetyltransferases

    • Cell cycle synchronization to account for cell cycle-dependent modifications

  • Temporal Controls:

    • Time-course experiments to capture dynamics (e.g., 0, 15, 30, 60, 120 minutes post-stimulus)

    • Parallel samples fixed at identical timepoints for cross-method validation

    • Reversibility assessment (stimulus addition/removal time course)

  • Cross-validation Controls:

    • Orthogonal detection methods (mass spectrometry, CUT&RUN)

    • Alternative antibodies targeting the same modification

    • ChIP-reChIP to assess co-occurrence with other modifications

The dynamic nature of histone modifications requires careful experimental design with appropriate temporal resolution and stimulus-specific positive controls to accurately capture the biological processes under investigation.

How can HIST1H3A (Ab-18) antibody be used in multiplexed histone modification assays?

Multiplexed assays allow simultaneous analysis of multiple histone modifications, providing comprehensive epigenetic profiles with minimal sample consumption. For integrating HIST1H3A (Ab-18) antibody into multiplexed approaches:

  • Luminex-Based Multiplex Histone PTM Assay:

    • HIST1H3A (Ab-18) antibody can be conjugated to color-coded magnetic beads

    • Combined with other histone PTM antibodies on different bead sets

    • Use biotinylated Histone H3 antibody as a reporter to recognize C-terminal domain

    • Detection via streptavidin-phycoerythrin signal

    • Normalization using Histone H3 Total antibody bead set

  • Mass Cytometry (CyTOF) Application:

    • Conjugate HIST1H3A (Ab-18) with rare earth metals

    • Combine with other metal-labeled histone antibodies

    • Enables single-cell analysis of multiple modifications

    • Requires optimization of antibody metal conjugation and staining protocols

  • Sequential Immunofluorescence:

    • Apply HIST1H3A (Ab-18) in first round of staining

    • Document signal and strip antibodies

    • Apply next antibody set and repeat

    • Computational alignment of images from sequential rounds

  • ChIP-Sequential Approach:

    • Perform initial ChIP with HIST1H3A (Ab-18)

    • Elute bound chromatin and perform second ChIP with different antibody

    • Repeat for multiple modifications

    • Reveals co-occurrence of modifications at specific genomic loci

These multiplexed approaches provide richer datasets than conventional single-antibody methods, enabling comprehensive analysis of the histone code with limited sample input .

What considerations are important when designing ChIP-seq experiments with this antibody?

ChIP-seq with HIST1H3A (Ab-18) antibody requires careful experimental design to generate high-quality, interpretable data:

  • Experimental Design Framework:

    • Include biological replicates (minimum 3)

    • Plan appropriate controls (input, IgG, spike-in)

    • Consider cell number requirements (typically 1-5×10⁶ cells per IP)

    • Design time-course or treatment conditions relevant to research question

  • Chromatin Preparation Optimization:

    • Fixation: 1% formaldehyde for 10 minutes at room temperature

    • Quenching: 125mM glycine for 5 minutes

    • Sonication: Optimize to achieve 200-500bp fragments

    • Verify fragmentation by agarose gel electrophoresis

  • Immunoprecipitation Parameters:

    • Antibody amount: 2-5μg per IP

    • Chromatin amount: 25-50μg per IP

    • Incubation: Overnight at 4°C with rotation

    • Bead selection: Protein A for rabbit polyclonal antibodies

    • Washing: Increase stringency progressively

  • Library Preparation Considerations:

    • Input normalization across samples

    • Size selection to remove adapter dimers

    • PCR cycle optimization to minimize amplification bias

    • Unique molecular identifiers (UMIs) to control for PCR duplicates

  • Data Analysis Strategy:

    • Peak calling algorithm selection (e.g., MACS2)

    • Normalization method (spike-in, input subtraction)

    • Differential binding analysis between conditions

    • Integration with other genomic data types (RNA-seq, ATAC-seq)

By addressing these considerations systematically, researchers can generate robust ChIP-seq data that accurately reflects the genomic distribution of histone H3 and its modifications.

How can HIST1H3A (Ab-18) antibody be used to investigate the relationship between histone modifications and gene expression?

Investigating the relationship between histone modifications and gene expression requires integrative experimental approaches. With HIST1H3A (Ab-18) antibody:

  • Integrated Genomics Approach:

    • ChIP-seq with HIST1H3A (Ab-18) to map genomic locations

    • RNA-seq on matched samples to correlate with gene expression

    • ATAC-seq to assess chromatin accessibility

    • Integration using computational methods to identify correlations

  • Single-Cell Multi-Omics:

    • scCUT&Tag with HIST1H3A (Ab-18) antibody

    • Paired with scRNA-seq on the same cells

    • Reveals cell-to-cell variation in histone modification-expression relationships

    • Requires specialized protocols for simultaneous profiling

  • Perturbation Studies:

    • Target writers/erasers of specific modifications using CRISPR or inhibitors

    • Monitor changes in both histone modifications (ChIP) and gene expression (RNA-seq)

    • Establish causality rather than correlation

    • Time-course designs can reveal kinetics of regulation

  • Regional Analysis Methodology:

    • Classify genomic regions (promoters, enhancers, gene bodies)

    • Correlate modification patterns with expression of associated genes

    • Conduct metagene analysis around transcription start sites

    • Identify cell type-specific regulatory elements

This integrated approach provides mechanistic insights into how specific histone modifications, including those recognized by HIST1H3A (Ab-18) antibody, contribute to transcriptional regulation in different genomic contexts and cellular states.

How can HIST1H3A (Ab-18) antibody be adapted for use in CUT&RUN or CUT&Tag protocols?

CUT&RUN and CUT&Tag represent significant advances over traditional ChIP for profiling histone modifications. For adapting HIST1H3A (Ab-18) antibody to these techniques:

CUT&RUN Protocol Adaptation:

  • Cell Preparation:

    • Use unfixed cells (200,000-500,000)

    • Bind to Concanavalin A-coated magnetic beads

    • Permeabilize with digitonin buffer

  • Antibody Parameters:

    • Dilute HIST1H3A (Ab-18) to 1:100-1:300

    • Incubate overnight at 4°C

    • Wash thoroughly to remove unbound antibody

  • pA-MNase Digestion:

    • Introduce protein A-MNase fusion protein

    • Activate with Ca²⁺ at optimal temperature (0-4°C)

    • Stop reaction with EGTA-containing buffer

  • Fragment Purification:

    • Extract released DNA fragments

    • Prepare sequencing libraries with low-input methods

    • Include spike-in controls for quantification

CUT&Tag Modifications:

  • Substitute pA-Tn5 transposase fusion protein for pA-MNase

  • Perform tagmentation directly on antibody-bound chromatin

  • Simplifies workflow and reduces processing time

Both techniques offer advantages over traditional ChIP-seq, including:

  • Reduced background signal

  • Lower cell number requirements

  • Higher resolution of binding sites

  • Improved signal-to-noise ratio

These methods are particularly valuable for limited samples and for detecting weakly enriched regions that might be missed by conventional ChIP-seq approaches.

What considerations are important when using HIST1H3A (Ab-18) antibody in single-cell epigenomics studies?

Single-cell epigenomic approaches present unique challenges for antibody-based histone modification detection. For HIST1H3A (Ab-18) antibody:

  • Technical Considerations:

    • Antibody specificity becomes even more critical at single-cell level

    • Signal amplification may be necessary due to limited material

    • Fixation protocols must balance epitope preservation with cell integrity

    • Background control is essential for reliable signal detection

  • Protocol Adaptations:

    • Reduce reaction volumes to maintain antibody concentration

    • Extend incubation times to ensure sufficient binding

    • Include carrier proteins/DNA to prevent non-specific loss

    • Optimize wash steps to remove background while preserving signal

  • Multi-modal Integration:

    • Design compatible workflows for simultaneous protein and DNA/RNA detection

    • Consider indexed sorting approaches for parallel processing

    • Develop computational methods to integrate multi-omic data

    • Account for technical variation in downstream analysis

  • Validation Strategies:

    • Benchmark against bulk population measurements

    • Use spike-in controls of known concentration

    • Perform dilution series to establish detection limits

    • Compare results from orthogonal single-cell methods

Single-cell approaches with HIST1H3A (Ab-18) antibody can reveal cellular heterogeneity in histone modification patterns that would be masked in bulk analysis, providing insights into cell state transitions and regulatory mechanisms at unprecedented resolution.

How does HIST1H3A (Ab-18) antibody performance compare in native versus cross-linked chromatin immunoprecipitation?

The choice between native and cross-linked ChIP significantly impacts experimental outcomes when using HIST1H3A (Ab-18) antibody:

Native ChIP (N-ChIP):

  • Maintains protein in natural state without chemical modification

  • Preserves epitopes that might be masked by formaldehyde

  • Typically yields higher signal-to-noise ratio for histone modifications

  • Limited to stable protein-DNA interactions

  • Requires careful handling to prevent modification loss

Cross-linked ChIP (X-ChIP):

  • Captures transient protein-DNA interactions

  • Preserves complex chromatin structures

  • Enables detection of proteins indirectly associated with chromatin

  • May reduce epitope accessibility due to crosslinking

  • Higher background signal is common

Comparative Performance Analysis:

ParameterNative ChIPCross-linked ChIP
Signal-to-noise ratioHigherLower
Epitope accessibilityBetter for histone coreBetter for transient factors
Fragment sizeTypically 150bp (mononucleosomes)Variable (200-500bp)
Protocol complexitySimplerMore complex
Preservation of complexesLimitedExcellent

Optimization Recommendations:

  • For histone core modifications: Native ChIP typically provides cleaner results

  • For studying complexes associated with H3: Cross-linked ChIP may be necessary

  • Perform side-by-side comparison with your specific biological system

  • Adjust antibody concentration based on protocol (typically 1.5-2× higher for X-ChIP)

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 2024 Thebiotek. All Rights Reserved.