HIST1H3A (Ab-10) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship your orders within 1-3 business days of receipt. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function
HIST1H3A is a core component of the nucleosome. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex interplay of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research indicates that epigenetic regulation in cancer is mediated by inducing E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Increased expression of H3K27me3 during a patient's clinical course may be helpful in determining whether tumors are heterochronous. PMID: 29482987
  3. Studies have shown that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease mediating histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Data suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) offers an alternative proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that in the early developing human brain, HIST1H3B comprises 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 frequently associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Research demonstrates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral particles, 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 describes, 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 associated with poor prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data show 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, while the H3K9me3 level was greatest during and immediately after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data suggest that the 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 the 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. Conversely, histone 3 mutations do not appear 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 Raji cells treated by staurosporin. PMID: 26032366
  28. 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) decrease 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 the incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) plays 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 the 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 are 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-10) Antibody and what specific target does it recognize?

HIST1H3A (Ab-10) Antibody is a polyclonal antibody raised in rabbits that specifically recognizes the peptide sequence surrounding Serine 10 of human Histone H3.1. Histone H3.1 is a fundamental component of the eukaryotic nucleosome, playing critical roles in chromatin structure and epigenetic regulation. The antibody specifically targets the region containing Serine 10, which is a key phosphorylation site involved in various cellular processes including mitosis and transcriptional activation . This antibody belongs to the IgG isotype and is typically available in unconjugated form for maximum flexibility in experimental applications .

What applications is HIST1H3A (Ab-10) Antibody validated for in research settings?

HIST1H3A (Ab-10) Antibody has been validated for multiple experimental applications crucial to epigenetic and chromatin research:

ApplicationRecommended DilutionSample TypesKey Considerations
Western Blot (WB)1:500-5000Cell acid extractsDetects ~17 kDa band
Immunohistochemistry (IHC)1:20-200Paraffin-embedded tissuesSuccessfully tested on human colon and lung cancer tissues
Immunofluorescence (IF)1:50-200Fixed cellsWorks well with HeLa cells
ELISAVariable by protocolPurified proteins/extractsCan detect nanogram quantities
Chromatin Immunoprecipitation (ChIP)2 μl per 500 μl solutionCrosslinked chromatinSuccessfully tested in HeLa cells treated with Micrococcal Nuclease

The antibody has shown specific reactivity in these applications with human samples, allowing researchers to study histone H3.1 modifications in various experimental contexts .

How is HIST1H3A (Ab-10) Antibody related to other histone H3 antibodies?

HIST1H3A (Ab-10) Antibody specifically targets histone H3.1 at the Serine 10 region, which distinguishes it from general histone H3 antibodies that may recognize multiple H3 variants. Histone H3 in humans exists in multiple variant forms including H3.1, H3.2, and H3.3, each encoded by different genes and having subtle differences in amino acid sequence and function . While general anti-H3 antibodies typically recognize conserved regions common to all variants, HIST1H3A (Ab-10) specifically targets the H3.1 variant with focus on the functionally important Ser10 region . This specificity makes it particularly valuable for studies examining phosphorylation-dependent processes at this site, which are associated with chromosome condensation during mitosis and transcriptional activation of immediate-early genes .

What are the optimal storage conditions for maintaining HIST1H3A (Ab-10) Antibody activity?

For optimal preservation of HIST1H3A (Ab-10) Antibody activity, the following storage protocol is recommended:

  • Short-term storage (up to 2 weeks): Maintain refrigerated at 2-8°C in its original buffer containing preservatives (typically 0.03% Proclin 300) and stabilizers (50% Glycerol) .

  • Long-term storage: Store at -20°C in small aliquots to prevent repeated freeze-thaw cycles that can denature antibody proteins and reduce binding efficacy .

  • Preparation before use: Briefly centrifuge tubes prior to opening to collect all material and avoid loss from adhesion to caps or tube walls .

  • Reconstitution (if lyophilized): Add the specified volume (typically 50 μl) of sterile water directly to the lyophilized powder and allow complete dissolution before use .

  • Working dilutions: Once diluted to working concentration, use immediately for best results or store briefly at 4°C .

The expected shelf life is approximately 12 months from the date of receipt when properly stored . Monitoring antibody performance in positive controls at regular intervals can help detect any loss of activity over time.

How should sample preparation be optimized for Western blot using HIST1H3A (Ab-10) Antibody?

For optimal Western blot results with HIST1H3A (Ab-10) Antibody, sample preparation is critical and should follow these methodological guidelines:

  • Cell Extraction Method: Use acid extraction techniques to efficiently isolate histones. This typically involves treating cells with a hypotonic lysis buffer followed by acid extraction (0.2N HCl) to solubilize histones .

  • Sample Types: The antibody has been validated with Jurkat and HEK293 cell acid extracts, which serve as appropriate positive controls .

  • Protein Loading: Load 10-20 μg of acid-extracted histones per lane for optimal detection.

  • Protein Transfer: Use PVDF membrane rather than nitrocellulose for better retention of small histone proteins.

  • Blocking Conditions: Block with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature.

  • Antibody Dilution: Use the antibody at 1:500-5000 dilution in blocking buffer .

  • Incubation Conditions: Incubate with primary antibody overnight at 4°C for best results.

  • Detection Method: HRP-conjugated secondary antibodies with enhanced chemiluminescence provide sensitive detection of the approximately 17 kDa histone H3.1 band .

  • Stripping and Reprobing: If necessary, mild stripping conditions should be used to avoid removing the small histone proteins from the membrane.

This optimized protocol enhances detection specificity and reduces background, particularly important when studying specific histone modifications in complex cellular extracts.

What are the critical parameters for successful ChIP experiments using HIST1H3A (Ab-10) Antibody?

Chromatin Immunoprecipitation (ChIP) using HIST1H3A (Ab-10) Antibody requires careful attention to several critical parameters for successful results:

  • Chromatin Preparation:

    • Crosslink cells optimally (typically 1% formaldehyde for 10 minutes at room temperature)

    • Treat with Micrococcal Nuclease to fragment chromatin to appropriate size (200-500 bp is ideal)

    • Sonication conditions should be optimized for each cell type to achieve consistent fragmentation

  • Immunoprecipitation Conditions:

    • Use 5 μg of HIST1H3A (Ab-10) antibody per ChIP reaction (approximately 4×10^6 cells)

    • Include appropriate negative controls (normal rabbit IgG at the same concentration)

    • Perform pre-clearing with protein A/G beads to reduce background

    • Use low-binding tubes to prevent antibody loss during incubation steps

  • Washing Stringency:

    • Progressive washing with increasing salt concentrations improves specificity

    • Include at least 4-5 wash steps with appropriate buffers to reduce non-specific binding

  • Elution and Crosslink Reversal:

    • Elute bound chromatin under gentle conditions to maintain DNA integrity

    • Reverse crosslinks completely (typically 65°C overnight)

    • Include RNase and Proteinase K treatments to remove contaminating RNA and protein

  • DNA Purification and Analysis:

    • Purify ChIP DNA using silica column-based methods for best recovery

    • Quantify DNA using real-time PCR with primers targeting known H3.1-associated genomic regions

    • Compare enrichment to input samples and IgG controls

Following these methodological guidelines enables accurate mapping of histone H3.1 distribution and specific modifications at Ser10 across the genome, providing insights into chromatin regulation during different cellular processes.

How can HIST1H3A (Ab-10) Antibody be used to investigate the relationship between histone phosphorylation and gene expression?

HIST1H3A (Ab-10) Antibody, targeting the Serine 10 region of histone H3.1, can be employed in sophisticated experimental designs to investigate the relationship between histone phosphorylation and gene expression:

  • Sequential ChIP (Re-ChIP) Analysis:

    • First immunoprecipitate with HIST1H3A (Ab-10) antibody

    • Then perform a second immunoprecipitation with antibodies against transcription factors or other histone modifications

    • This reveals genomic loci where H3S10 phosphorylation co-occurs with specific transcriptional regulators

  • ChIP-seq with Stimulus-Response Design:

    • Perform ChIP-seq using HIST1H3A (Ab-10) antibody before and after cell stimulation (e.g., growth factors, stress, mitogenic signals)

    • Map genome-wide changes in H3S10 phosphorylation patterns

    • Correlate with RNA-seq data from matched samples to directly link phosphorylation events with transcriptional outcomes

  • Pharmacological Inhibitor Studies:

    • Treat cells with specific kinase inhibitors (e.g., Aurora B, MSK1/2, RSK inhibitors)

    • Assess changes in H3S10 phosphorylation using the antibody in Western blot or ChIP assays

    • Correlate with changes in expression of immediate-early genes to establish causative relationships

  • Time-Course Immunofluorescence:

    • Use the antibody at 1:50-200 dilution for immunofluorescence

    • Perform time-course experiments after stimulation to visualize the dynamics of H3S10 phosphorylation

    • Co-stain with RNA Polymerase II phospho-antibodies to correlate with transcriptional activation

  • Cell-Cycle Synchronization Studies:

    • Synchronize cells at different cell cycle phases

    • Use the antibody to track H3S10 phosphorylation during mitosis and interphase

    • Correlate with expression of cell-cycle regulated genes

These methodological approaches provide mechanistic insights into how H3S10 phosphorylation contributes to chromatin remodeling and transcriptional regulation in various biological contexts, from cell division to stimulus-responsive gene expression programs.

What are common troubleshooting strategies for weak or non-specific signal when using HIST1H3A (Ab-10) Antibody?

When encountering weak or non-specific signals with HIST1H3A (Ab-10) Antibody, the following methodological troubleshooting strategies can help optimize experimental outcomes:

IssuePossible CausesTroubleshooting Strategies
Weak Signal in Western Blot- Insufficient antigen amount
- Incomplete protein transfer
- Antibody dilution too high
- Inadequate exposure time
- Increase protein loading (20-40 μg of acid-extracted histones)
- Use transfer conditions optimized for small proteins (wet transfer)
- Decrease antibody dilution to 1:500-1:1000
- Increase exposure time or use more sensitive detection reagents
High Background in Western Blot- Insufficient blocking
- Insufficient washing
- Antibody concentration too high
- Extend blocking time to 2 hours
- Use 5% BSA instead of milk for blocking
- Increase washing steps (5-6 times for 5-10 minutes each)
- Increase antibody dilution to 1:2000-1:5000
Poor Signal in IHC/IF- Inadequate antigen retrieval
- Fixation issues
- Suboptimal antibody incubation
- Optimize antigen retrieval (try citrate buffer pH 6.0 with heat)
- Test different fixation methods (4% PFA vs. methanol)
- Extend primary antibody incubation to overnight at 4°C
- Use signal amplification systems
Non-specific Bands in Western Blot- Cross-reactivity with other histone variants
- Degradation products
- Post-translational modifications
- Include peptide competition controls
- Use freshly prepared samples with protease inhibitors
- Compare with other validated H3 antibodies
- Run a histone H3.1 recombinant protein as positive control
Poor ChIP Efficiency- Insufficient chromatin fragmentation
- Suboptimal crosslinking
- Insufficient antibody amount
- Excessive washing stringency
- Optimize sonication/MNase digestion conditions
- Test different crosslinking times (8-12 minutes)
- Increase antibody amount to 5-10 μg per reaction
- Adjust salt concentration in wash buffers

Implementing these methodological refinements according to the specific experimental context can significantly improve both signal specificity and sensitivity when working with HIST1H3A (Ab-10) Antibody across different applications.

How can HIST1H3A (Ab-10) Antibody be used in multiplex immunofluorescence to study chromatin dynamics?

HIST1H3A (Ab-10) Antibody can be employed in sophisticated multiplex immunofluorescence strategies to investigate chromatin dynamics in various biological contexts:

These methodological approaches enable researchers to visualize and quantify the spatial and temporal dynamics of H3S10 phosphorylation in relation to other nuclear events, providing insights into chromatin reorganization during processes such as transcriptional activation, cell division, and cellular stress responses.

How does HIST1H3A (Ab-10) Antibody performance compare with other histone H3 antibodies in epigenetic research?

When designing epigenetic research studies, understanding the comparative advantages and limitations of HIST1H3A (Ab-10) Antibody versus other histone H3 antibodies is essential:

Antibody TypeTarget SpecificityKey ApplicationsAdvantagesLimitations
HIST1H3A (Ab-10)H3.1 variant with focus on Ser10 regionChIP, WB, IHC, IF, ELISA - High specificity for H3.1 Ser10 region
- Ideal for phosphorylation studies
- Well-validated in multiple applications
- Limited to human samples
- Polyclonal nature may introduce batch variation
General Histone H3Conserved regions of all H3 variantsAll standard applications, marker for nuclear compartment - Broader reactivity across species
- Useful as loading control
- Can detect all H3 variants
- Cannot distinguish between H3 variants
- Less useful for specific modification studies
H3 Phospho-Specific (pS10)Phosphorylated Ser10 epitopeCell cycle studies, mitosis markers- Direct measurement of phosphorylation state
- High specificity for active mark
- May not distinguish between H3 variants
- Sensitive to phosphatase activity in samples
H3 Variant-Specific (H3.3)Regions unique to H3.3Transcription studies, ChIP-seq- Distinguishes replacement variant
- Useful for active chromatin studies
- Not appropriate for cell cycle studies
- May not focus on modification sites

Methodological considerations for comparative studies:

  • Sequential or Parallel ChIP Analysis:

    • Using HIST1H3A (Ab-10) in parallel with other H3 antibodies on identical samples

    • Comparing enrichment profiles to distinguish variant-specific from pan-H3 patterns

  • Cross-Validation Strategy:

    • Confirming key findings with multiple antibodies targeting different epitopes of H3

    • Using recombinant H3.1 versus H3.3 proteins as controls for specificity testing

  • Application-Specific Selection Criteria:

    • For studies focusing on mitosis and cell cycle: HIST1H3A (Ab-10) provides advantages due to its Ser10 region recognition

    • For broad chromatin occupancy studies: general H3 antibodies may provide more comprehensive coverage

    • For studies requiring cross-species comparison: broader reactivity antibodies like Agrisera's Anti-H3 may be more suitable

This comparative approach ensures appropriate antibody selection based on specific research questions and experimental designs in epigenetic studies.

What experimental controls should be included when using HIST1H3A (Ab-10) Antibody for phosphorylation-specific studies?

When conducting phosphorylation-specific studies using HIST1H3A (Ab-10) Antibody, rigorous experimental controls are essential for valid data interpretation:

  • Positive Controls:

    • Mitotic cell extracts (nocodazole-arrested) known to have high levels of H3S10 phosphorylation

    • Stimulated cells treated with agents that activate MAP kinase pathways (e.g., TPA, EGF)

    • Recombinant H3.1 protein pre-phosphorylated at Ser10 by Aurora B kinase in vitro

  • Negative Controls:

    • Phosphatase-treated samples to remove phosphorylation at Ser10

    • Cells treated with kinase inhibitors specific for H3S10 kinases (Aurora B, MSK1/2)

    • Non-phosphorylatable H3.1 mutant (S10A) expressed in cells

  • Specificity Controls:

    • Peptide competition assays using:

      • Phosphorylated H3S10 peptide (should block signal)

      • Non-phosphorylated H3S10 peptide (should not block signal)

      • Phosphorylated peptides from other sites (H3S28ph - should not block)

  • Antibody Validation Controls:

    • Comparison with commercially available phospho-specific anti-H3S10ph antibodies

    • Immunoprecipitation followed by mass spectrometry to confirm target identity

    • Sequential probing with general H3 antibody after stripping to confirm protein loading

  • Technical Controls for ChIP Experiments:

    • Input chromatin (non-immunoprecipitated) as reference

    • Non-specific IgG from the same species (rabbit) as negative control

    • ChIP using general H3 antibody to normalize for nucleosome occupancy

    • Known genomic regions with constitutive or inducible H3S10 phosphorylation as positive loci

  • Treatment Validation:

    • Parallel Western blot analysis to confirm phosphorylation status changes

    • Immunofluorescence to visualize cellular distribution of signals

    • Time-course analysis to capture dynamic phosphorylation events

Implementing this comprehensive control strategy ensures that observed signals genuinely represent H3.1 Ser10 phosphorylation events, enabling confident interpretation of experimental results in studies investigating chromatin regulation, cell cycle progression, or transcriptional activation.

How should researchers design experiments to distinguish between different histone H3 variants using HIST1H3A (Ab-10) Antibody?

Designing experiments to effectively distinguish between different histone H3 variants using HIST1H3A (Ab-10) Antibody requires strategic approaches that leverage the antibody's specificity for H3.1 while implementing controls to differentiate from other H3 variants:

  • Sequential Immunoprecipitation Protocol:

    • First round: Immunoprecipitate with general H3 antibody

    • Split the sample and perform second round with:

      • HIST1H3A (Ab-10) Antibody to enrich for H3.1

      • H3.3-specific antibody to enrich for H3.3

    • Compare genomic distributions to identify variant-specific regions

  • Comparative ChIP-seq Experimental Design:

    • Perform parallel ChIP-seq with:

      • HIST1H3A (Ab-10) Antibody (H3.1-enriched)

      • H3.3-specific antibody

      • Pan-H3 antibody

    • Bioinformatic analysis to identify:

      • H3.1-specific regions (enriched with HIST1H3A but not H3.3)

      • H3.3-specific regions (enriched with H3.3 but not HIST1H3A)

      • Common regions (enriched with both antibodies)

  • Cell Cycle-Dependent Analysis:

    • Synchronize cells at different cell cycle phases:

      • G1/G0 (low H3.1 incorporation)

      • S phase (high H3.1 incorporation)

      • G2/M (high H3S10 phosphorylation)

    • Compare HIST1H3A (Ab-10) signals across these phases by Western blot and IF

    • Correlate with replication timing and transcriptional activity

  • Genetic Knockdown Validation Strategy:

    • Perform siRNA-mediated knockdown of:

      • H3.1-specific chaperone (CAF-1)

      • H3.3-specific chaperone (HIRA, DAXX)

    • Assess changes in HIST1H3A (Ab-10) signal distribution

    • Validate with complementary antibodies and quantitative PCR

  • Exogenous Expression System:

    • Express tagged versions of histone variants:

      • FLAG-H3.1

      • HA-H3.3

    • Perform ChIP with:

      • HIST1H3A (Ab-10) Antibody

      • Anti-FLAG (H3.1-specific)

      • Anti-HA (H3.3-specific)

    • Compare enrichment profiles to validate variant specificity

  • Pulse-Chase Experimental Design:

    • Label newly synthesized histones (e.g., SNAP-tag approaches)

    • Track incorporation dynamics over time

    • Correlate with HIST1H3A (Ab-10) signals to distinguish replication-dependent (H3.1) from replication-independent (H3.3) deposition

These methodological approaches enable researchers to effectively distinguish between histone H3 variants in various biological contexts, providing insights into their distinct functions in chromatin regulation, DNA replication, transcription, and cell division processes.

How can HIST1H3A (Ab-10) Antibody be integrated into single-cell epigenomic analyses?

The integration of HIST1H3A (Ab-10) Antibody into emerging single-cell epigenomic technologies represents a frontier in chromatin research methodology:

  • Single-Cell CUT&Tag Protocol Adaptation:

    • Optimize HIST1H3A (Ab-10) Antibody concentration for limited cell numbers

    • Develop a protocol utilizing protein A-Tn5 transposase fusion proteins with HIST1H3A (Ab-10)

    • Implement cell sorting to isolate specific populations prior to analysis

    • Develop computational pipelines to analyze H3.1 Ser10 phosphorylation patterns at single-cell resolution

  • Antibody-Based Single-Cell Combinatorial Indexing:

    • Use HIST1H3A (Ab-10) in combination with antibodies against other chromatin features

    • Implement split-pool barcoding strategies to profile thousands of individual cells

    • Create multidimensional maps of H3.1 distribution and modification states across heterogeneous populations

  • Integrated Multi-Omics Approaches:

    • Develop protocols combining:

      • Single-cell H3.1 ChIP-seq using HIST1H3A (Ab-10)

      • Single-cell RNA-seq on the same cells

      • Single-cell ATAC-seq for chromatin accessibility

    • Correlate H3.1 occupancy with gene expression and chromatin openness at single-cell level

  • Microfluidic Platform Integration:

    • Adapt HIST1H3A (Ab-10) ChIP protocols to microfluidic devices

    • Minimize reagent volumes and maximize sensitivity for single-cell applications

    • Implement on-chip washing and purification steps to reduce background

  • Spatial Epigenomics Applications:

    • Combine HIST1H3A (Ab-10) immunofluorescence with in situ sequencing

    • Develop protocols for spatially resolved ChIP-seq using HIST1H3A (Ab-10)

    • Map H3.1 distribution in tissue contexts while preserving spatial information

  • Advanced Data Analysis Frameworks:

    • Develop computational methods to account for technical variation in single-cell epigenomic data

    • Implement trajectory analysis to map H3.1 dynamics during cellular differentiation or response

    • Create integrative models connecting H3.1 phosphorylation states to transcriptional outcomes

These emerging methodologies will enable unprecedented insights into the cell-to-cell variation in histone H3.1 distribution and modification, revealing how epigenetic heterogeneity contributes to cellular function and disease processes at single-cell resolution.

What are the challenges and potential solutions for using HIST1H3A (Ab-10) Antibody in long-term time-lapse experiments?

Long-term time-lapse experiments using HIST1H3A (Ab-10) Antibody present specific challenges that require methodological solutions:

  • Challenge: Antibody Accessibility in Living Cells

    • Solutions:

      • Develop cell-permeable versions of HIST1H3A (Ab-10) using protein transduction domains

      • Create Fab fragments with improved cellular penetration while maintaining specificity

      • Implement reversible permeabilization protocols that allow antibody entry while maintaining cell viability

  • Challenge: Phototoxicity and Photobleaching

    • Solutions:

      • Use photostable fluorophores (e.g., Alexa Fluor 647) for antibody conjugation

      • Implement intelligent illumination strategies (minimal exposure, optimized intervals)

      • Develop oxygen scavenging systems to reduce photobleaching during long-term imaging

  • Challenge: Maintaining Physiological Conditions

    • Solutions:

      • Optimize imaging media composition to support long-term cell health

      • Implement environmental control systems (temperature, CO2, humidity)

      • Develop microfluidic chambers for continuous media exchange without disturbing imaging

  • Challenge: Signal-to-Noise Ratio Degradation Over Time

    • Solutions:

      • Apply denoising algorithms specifically designed for time-lapse data

      • Implement signal amplification strategies (e.g., tyramide signal amplification)

      • Develop computational approaches to correct for background increase and signal decay

  • Challenge: Tracking Individual Cells Through Division

    • Solutions:

      • Combine HIST1H3A (Ab-10) with stable cell tracking markers (nuclear localized fluorescent proteins)

      • Implement machine learning-based cell tracking algorithms

      • Develop lineage reconstruction methods that account for mitotic events

  • Challenge: Quantifying Dynamic Phosphorylation Changes

    • Solutions:

      • Create ratiometric imaging approaches using co-staining with total H3 antibodies

      • Implement FRET-based sensors to detect H3S10 phosphorylation in real-time

      • Develop calibration standards for quantitative interpretation of fluorescence intensity

  • Challenge: Correlating with Functional Outcomes

    • Solutions:

      • Design dual-reporter systems that simultaneously track H3S10 phosphorylation and transcriptional activity

      • Implement post-experiment fixation and staining protocols for endpoint analysis

      • Develop computational frameworks to correlate dynamic histone modifications with cellular behaviors

These methodological approaches address the technical challenges of using HIST1H3A (Ab-10) Antibody in long-term live-cell imaging experiments, enabling researchers to visualize and quantify the dynamics of H3.1 phosphorylation in relation to cell cycle progression, transcriptional regulation, and response to environmental stimuli over extended time periods.

What standardized quality control procedures should researchers implement when working with HIST1H3A (Ab-10) Antibody?

To ensure experimental reproducibility and valid data interpretation when working with HIST1H3A (Ab-10) Antibody, researchers should implement the following standardized quality control procedures:

  • Antibody Validation Protocol:

    • Western blot validation using:

      • Positive controls (HEK293 or Jurkat acid extracts)

      • Negative controls (non-human samples or phosphatase-treated extracts)

      • Peptide competition assays with phosphorylated and non-phosphorylated peptides

    • Lot-to-lot validation when receiving new antibody batches

    • Regular testing with positive controls to monitor antibody performance over time

  • Application-Specific Quality Controls:

    • For Western Blot:

      • Include molecular weight markers to confirm 17 kDa band position

      • Use loading controls (total protein stain or stable housekeeping proteins)

      • Include recombinant H3.1 protein as reference standard

    • For ChIP/ChIP-seq:

      • Assess chromatin fragmentation quality by gel electrophoresis

      • Include spike-in controls for normalization

      • Validate enrichment at known positive and negative loci by qPCR

      • Calculate signal-to-noise ratio and fragment size distribution metrics

    • For Immunofluorescence:

      • Include positive cell populations (mitotic cells)

      • Use blocking peptides to confirm signal specificity

      • Implement no-primary antibody controls

  • Standardized Reporting Requirements:

    • Document key experimental parameters:

      • Antibody catalog number, lot number, and dilution used

      • Detailed sample preparation methods

      • Incubation times and temperatures

      • Buffer compositions

      • Image acquisition settings

    • Maintain detailed laboratory records of antibody performance across experiments

  • Periodic Performance Assessment:

    • Test antibody sensitivity using dilution series of positive control samples

    • Assess specificity through immunoprecipitation followed by mass spectrometry

    • Compare performance with alternative antibodies targeting the same epitope

  • Collaborative Validation:

    • Participate in antibody validation initiatives

    • Share validation data through repositories or publications

    • Implement multi-laboratory testing for critical experiments

What emerging technologies might enhance or replace the use of HIST1H3A (Ab-10) Antibody in future epigenetic research?

The landscape of epigenetic research tools is rapidly evolving, with several emerging technologies poised to complement or potentially replace traditional antibody-based approaches like HIST1H3A (Ab-10) Antibody:

  • Engineered Protein-Based Detection Systems:

    • CRISPR-based histone modification detectors

    • Synthetic histone modification reader domains with fluorescent outputs

    • Nanobodies with enhanced specificity for histone H3.1 and its modifications

    • Split protein complementation systems for live-cell visualization of H3S10 phosphorylation

  • Mass Spectrometry Advancements:

    • Targeted mass spectrometry approaches for precise quantification of H3.1 phosphorylation

    • Single-cell proteomics methods for analyzing histone modifications

    • Imaging mass spectrometry for spatial mapping of histone variants

    • Top-down proteomics for analyzing combinatorial histone modification patterns

  • Genomic and Sequencing Technologies:

    • CUT&Tag methods optimized for histone variant-specific mapping

    • Direct detection of histone modifications during nanopore sequencing

    • Long-read sequencing approaches for linking distant epigenetic features

    • Combinatorial indexing methods for ultra-high-throughput single-cell epigenomics

  • Synthetic Biology Approaches:

    • Engineered cells with endogenously tagged histone H3.1

    • Orthogonal histone-DNA interaction systems for tracking specific nucleosome populations

    • Synthetic histone modification circuits with programmable outputs

    • Bio-orthogonal chemistry for specific labeling of newly synthesized histones

  • Advanced Imaging Methodologies:

    • Super-resolution microscopy optimized for chromatin architecture

    • Lattice light-sheet microscopy for long-term 3D imaging of histone dynamics

    • Expansion microscopy for enhanced visualization of nuclear subcompartments

    • Label-free imaging methods that detect biophysical properties of modified chromatin

  • Computational and AI-Enhanced Analysis:

    • Deep learning approaches for identifying histone modification patterns

    • Integrative multi-omics frameworks connecting histone states to functional outcomes

    • Predictive modeling of histone modification dynamics based on cellular contexts

    • Automated image analysis pipelines for high-content screening of chromatin states

While these emerging technologies may eventually reduce reliance on traditional antibodies like HIST1H3A (Ab-10), the immediate future will likely involve hybrid approaches that combine antibody-based detection with these advanced technologies to provide complementary data streams and validation. The integration of multiple methodologies will drive more comprehensive understanding of histone H3.1 biology and its role in chromatin regulation.

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.