HIST1H3A (Ab-17) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

HIST1H3A (Ab-17) Antibody is a rabbit-derived polyclonal IgG antibody that recognizes the histone H3.1 variant encoded by the HIST1H3A gene. It targets a peptide sequence surrounding the unmodified arginine 17 (R17) residue of human histone H3.1, as specified by its immunogen design . This antibody is part of a broader toolkit for studying histone localization, chromatin remodeling, and post-translational modifications (PTMs) .

PropertyDetail
Target EpitopeArginine 17 (R17) on HIST1H3A (UniProt ID: P68431)
Host SpeciesRabbit
ClonalityPolyclonal
ApplicationsELISA, Immunofluorescence (IF), Immunohistochemistry (IHC)

Key Use Cases

  • Chromatin Immunoprecipitation (ChIP): Identifies H3.1 occupancy in gene regulatory regions .

  • Cancer Research: Used in immunohistochemistry (IHC) to study histone localization in colon cancer tissues .

  • Cellular Imaging: Detects nuclear H3.1 in HeLa cells via immunofluorescence (IF) .

Validation Data

  • Western Blot: Detects a ~15 kDa band in human cell lysates (e.g., 293T, HeLa) .

  • IHC-Paraffin: Shows strong nuclear staining in formalin-fixed, paraffin-embedded tissues at dilutions of 1:20–1:200 .

Epigenetic Regulation

  • H3.1 deposition correlates with DNA replication and heterochromatin maintenance, contrasting with H3.3’s role in active transcription .

  • Aberrant H3.1 localization is implicated in cancers, such as gliomas, where histone mutations disrupt chromatin architecture .

Disease Associations

  • Cancer: Overexpression or mislocalization of H3.1 is observed in colorectal and cervical cancers .

  • Neurodevelopmental Disorders: Mutations in H3.1 are linked to developmental defects through disrupted Polycomb repressive complex (PRC) activity .

Comparative Analysis of HAntibodies

AntibodyTarget ModificationApplicationsClonality
HIST1H3A (Ab-17) Unmodified R17IHC, IF, ELISAPolyclonal
Mono-methyl-H3.1 (R17) R17 mono-methylationWB, ICC, IFMonoclonal
Symmetric di-methyl R17 R17 di-methylationWB, PepArr, ICC/IFMonoclonal

Limitations and Considerations

  • Modification-Specificity: Does not distinguish between modified (e.g., methylated, citrullinated) and unmodified R17 .

  • Species Restriction: Limited to human samples unless cross-validated .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method and location. For specific delivery timeframes, please consult your local distributors.
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
Histone H3 is a core component of nucleosomes. Nucleosomes package and condense DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones, therefore, play a pivotal 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, collectively known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer is mediated by the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The detection of increased expression of H3K27me3 during a patient's clinical course can aid in determining whether the tumors are heterochronous. PMID: 29482987
  3. Research has shown that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Data indicates that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) is 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 research demonstrates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This study of 47 diffuse midline gliomas revealed that 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. Among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Data suggests that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral DNA, and deposits histone H3.3 onto these. PMID: 28981850
  9. Experiments have demonstrated 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 provides the first description of 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 prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. The presence of H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data suggests 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 immediately after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Research indicates 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 efficiently control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggests 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 genuine 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 indicates 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 suggests 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 indicates 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 indicates that lower-resolution mass spectrometry instruments can be utilized for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data suggests 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-17) antibody and what epitope does it recognize?

HIST1H3A (Ab-17) antibody is a rabbit polyclonal antibody that specifically recognizes the peptide sequence around the arginine 17 (R17) site of human Histone H3.1. It is an unconjugated IgG antibody developed for detecting the HIST1H3A protein in various experimental applications. This antibody serves as a valuable tool for studying histone modifications and epigenetic regulation mechanisms .

The antibody's specificity for the R17 region is particularly important because this site undergoes post-translational modifications such as methylation, which plays crucial roles in transcriptional regulation, chromatin structure maintenance, and epigenetic signaling pathways .

What are the validated applications for HIST1H3A (Ab-17) antibody?

HIST1H3A (Ab-17) antibody has been validated for multiple experimental applications, providing researchers with versatility in their investigations. The primary validated applications include:

ApplicationRecommended DilutionValidated Cell/Tissue Types
ELISA1:500-1:2000Multiple human cell lines
Immunofluorescence (IF)1:50-1:200HeLa cells
Immunohistochemistry (IHC)1:20-1:200Human colon cancer tissue

The antibody demonstrates robust signal-to-noise ratio when used within these recommended dilution ranges. For novel applications or cell types, optimization of antibody concentration is advisable to achieve optimal results .

How should HIST1H3A (Ab-17) antibody be stored to maintain its activity?

Proper storage of HIST1H3A (Ab-17) antibody is critical for maintaining its activity and specificity. The manufacturer recommends the following storage conditions:

  • Short-term storage (up to 2 weeks): Maintain refrigerated at 2-8°C

  • Long-term storage: Store at -20°C in small aliquots to prevent freeze-thaw cycles

The antibody is supplied in a buffer containing 0.03% Proclin 300 and 50% glycerol, which helps maintain stability during storage. When handled correctly, the antibody remains active for up to 12 months from the date of receipt .

To minimize activity loss, avoid repeated freeze-thaw cycles by preparing appropriate working aliquots during initial thawing. Each freeze-thaw cycle can result in approximately 10-15% loss of antibody activity.

How can I optimize HIST1H3A (Ab-17) antibody for immunofluorescence experiments?

Optimizing HIST1H3A (Ab-17) antibody for immunofluorescence requires careful consideration of several experimental parameters:

  • Fixation method: 4% paraformaldehyde for 10-15 minutes at room temperature is recommended for preserving histone epitopes while maintaining cellular architecture.

  • Permeabilization: Use 0.1-0.5% Triton X-100 for 5-10 minutes to allow antibody access to nuclear antigens. Excessive permeabilization may disrupt nuclear structure and result in non-specific binding.

  • Blocking conditions: 5% normal serum (from the species of secondary antibody) in PBS with 0.1% Tween-20 for 1 hour at room temperature effectively reduces background.

  • Antibody dilution: Start with 1:100 dilution and adjust based on signal intensity. The recommended range is 1:50-1:200 .

  • Incubation conditions: Overnight incubation at 4°C typically yields optimal results with minimal background.

  • Controls: Include a negative control (no primary antibody) and positive control (known positive samples, such as HeLa cells) in each experiment .

The antibody has been successfully used for immunofluorescent analysis of HeLa cells, demonstrating clear nuclear localization patterns consistent with histone H3.1 distribution .

What are the critical factors for successful immunohistochemistry using HIST1H3A (Ab-17) antibody?

For successful immunohistochemistry using HIST1H3A (Ab-17) antibody, researchers should consider these critical factors:

  • Tissue preparation: Optimal fixation with 10% neutral buffered formalin for 24-48 hours, followed by paraffin embedding and sectioning at 4-6 μm thickness.

  • Antigen retrieval: Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) for 15-20 minutes is essential for exposing the R17 epitope that may be masked during fixation.

  • Endogenous peroxidase blocking: Treat sections with 3% hydrogen peroxide in methanol for 10-15 minutes to prevent non-specific background if using HRP-conjugated detection systems.

  • Antibody dilution range: The recommended dilution range is 1:20-1:200. Start with 1:50 and adjust based on staining intensity and background levels .

  • Incubation time and temperature: Overnight incubation at 4°C generally provides the best balance between specific signal and background.

  • Detection system: A polymer-based detection system often provides better sensitivity than biotin-streptavidin methods for histone modifications.

  • Counterstaining: Light hematoxylin counterstaining preserves visibility of nuclear staining patterns.

The antibody has demonstrated specific nuclear staining in paraffin-embedded human colon cancer tissue samples, with variations in staining intensity correlating with different patterns of histone H3.1 expression and modification .

How can I validate the specificity of HIST1H3A (Ab-17) antibody in my experimental system?

Validating antibody specificity is critical for reliable experimental results. For HIST1H3A (Ab-17) antibody, consider implementing these validation strategies:

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide (sequence around R17 of Histone H3.1) prior to application. This should abolish specific staining if the antibody is truly specific.

  • Western blot analysis: The antibody should detect a single band at approximately 15-17 kDa corresponding to Histone H3.1 .

  • Knockout/knockdown controls: Compare staining in wild-type cells versus cells where HIST1H3A expression has been reduced through siRNA or CRISPR-Cas9 techniques.

  • Cross-reactivity assessment: Test the antibody on tissues/cells from different species. The antibody is specifically designed for human samples, so minimal reactivity with mouse or rat samples would support its specificity .

  • Comparison with other established Histone H3 antibodies: Parallel staining with well-characterized H3 antibodies should show similar nuclear localization patterns but potentially different intensities depending on the epitope modification state.

  • Mass spectrometry correlation: For advanced validation, correlate antibody-based detection with mass spectrometry analysis of histone modifications at the R17 position.

Implementing multiple validation approaches provides stronger evidence for antibody specificity than relying on a single method.

How can HIST1H3A (Ab-17) antibody be utilized in chromatin immunoprecipitation (ChIP) experiments?

While HIST1H3A (Ab-17) antibody is not explicitly listed for ChIP in the product information, similar Histone H3 antibodies have demonstrated effectiveness in ChIP applications . For adapting this antibody to ChIP protocols:

  • Crosslinking optimization: Use 1% formaldehyde for 10 minutes at room temperature for protein-DNA crosslinking. Over-fixation can mask epitopes and reduce immunoprecipitation efficiency.

  • Chromatin fragmentation: Sonicate to achieve fragments of 200-500 bp for optimal immunoprecipitation. Confirm fragmentation by agarose gel electrophoresis.

  • Antibody amount: Start with 3-5 μg of antibody per ChIP reaction, adjusting based on preliminary results.

  • Beads selection: Protein A/G magnetic beads typically perform well with rabbit IgG antibodies.

  • Washing stringency: Include high-salt washes (up to 500 mM NaCl) to reduce non-specific binding while preserving specific interactions.

  • Elution conditions: Use gentle elution conditions (SDS and heat) to release antibody-bound chromatin complexes without denaturing the antibody.

  • Controls: Include IgG control, input control, and positive control (antibody against a known abundant histone mark) in each experiment.

When analyzing ChIP-seq data from HIST1H3A (Ab-17) antibody experiments, focus on genomic regions known to be regulated by modifications at R17, such as promoters of genes involved in cell cycle regulation and developmental pathways .

How does methylation at R17 affect epitope recognition by HIST1H3A (Ab-17) antibody?

The HIST1H3A (Ab-17) antibody recognizes the region surrounding arginine 17 (R17) on Histone H3.1. When using this antibody, it's crucial to understand how post-translational modifications affect epitope recognition:

  • Unmodified R17: The antibody shows highest affinity for the unmodified R17 epitope.

  • Mono-methylated R17: Mono-methylation at R17 significantly reduces antibody binding, as indicated by the existence of specific antibodies designed to recognize this modification .

  • Dimethylated R17: This modification likely prevents antibody binding completely.

  • Nearby modifications: Modifications at nearby residues (K14, K18) may also influence antibody recognition.

This differential recognition has important research implications:

  • When studying global Histone H3.1 distribution, be aware that regions with high R17 methylation might be underrepresented.

  • For comprehensive analysis of Histone H3.1 regardless of modification state, consider using antibodies targeting more conserved regions or unmodifiable residues.

  • When unexpected staining patterns emerge, consider the possibility of differential R17 modification across cell types or conditions.

Researchers studying the dynamics of R17 methylation specifically would benefit from using modification-specific antibodies like the mono-methyl-Histone H3.1 (R17) antibody in parallel with the HIST1H3A (Ab-17) antibody.

How can HIST1H3A (Ab-17) antibody be used in multiplexed immunofluorescence studies?

Multiplexed immunofluorescence allows simultaneous detection of multiple targets, providing valuable insights into the spatial relationships between different proteins or modifications. For incorporating HIST1H3A (Ab-17) antibody into multiplexed studies:

  • Antibody compatibility planning:

    • Choose companion antibodies from different host species to avoid cross-reactivity

    • If using multiple rabbit antibodies, consider sequential staining with direct labeling or tyramide signal amplification (TSA)

    • The antibody works well with mouse anti-tubulin antibodies for co-localization studies

  • Optimized detection strategy:

    • Recommended secondary antibody: Anti-rabbit IgG conjugated to Alexa Fluor 488, 568, or 647

    • Starting dilution for secondary antibody: 1:500-1:1000

    • Consider using nuclear counterstains that don't interfere with histone visualization (DAPI at 1:10,000 works well)

  • Potential multiplexing combinations:

    Primary Antibody 1Primary Antibody 2Primary Antibody 3Application
    HIST1H3A (Ab-17)Mouse anti-H3K9me3Goat anti-RNA Pol IITranscriptional repression studies
    HIST1H3A (Ab-17)Mouse anti-γH2AXGoat anti-53BP1DNA damage response analysis
    HIST1H3A (Ab-17)Mouse anti-Ki67Goat anti-cleaved caspase-3Cell cycle/apoptosis relationship
  • Sequential staining protocol:

    • Apply HIST1H3A (Ab-17) antibody first (1:100 dilution)

    • Detect with secondary antibody

    • Apply fixative (4% PFA, 10 minutes) to prevent antibody dissociation

    • Proceed with second primary antibody

    • Repeat for third antibody if applicable

  • Spectral unmixing: For advanced confocal microscopy, use spectral unmixing to separate overlapping fluorophore signals, especially important when studying nuclear proteins with similar localization patterns.

This approach enables complex analyses such as correlating H3.1 distribution with specific histone modifications or transcriptional regulators in the same cellular compartments .

What are common issues with HIST1H3A (Ab-17) antibody in immunohistochemistry and how can they be resolved?

Researchers may encounter several challenges when using HIST1H3A (Ab-17) antibody for immunohistochemistry. Here are common issues and their solutions:

  • Weak or absent signal:

    • Cause: Inadequate antigen retrieval, overfixation, or antibody concentration too low

    • Solution: Extend antigen retrieval time to 25-30 minutes, optimize fixation time (24-48 hours), or increase antibody concentration (try 1:20 dilution)

  • High background staining:

    • Cause: Insufficient blocking, antibody concentration too high, or non-specific binding

    • Solution: Extend blocking time to 2 hours, dilute antibody further (1:200), or add 0.1% Tween-20 to washing buffers

  • Cytoplasmic rather than nuclear staining:

    • Cause: Possible cross-reactivity or cell fixation issues

    • Solution: Reduce primary antibody incubation time, optimize fixation protocol, or perform peptide competition assay to confirm specificity

  • Inconsistent staining intensity across the tissue section:

    • Cause: Uneven antigen retrieval or antibody application

    • Solution: Ensure complete tissue immersion during antigen retrieval and apply sufficient volume of antibody solution

  • Signal in negative control sections:

    • Cause: Endogenous peroxidase activity or non-specific binding of secondary antibody

    • Solution: Enhance endogenous peroxidase blocking (3% H2O2 for 20 minutes) or use more stringent washing conditions

Quality control steps to implement:

  • Include positive control tissue (human colon cancer) in each staining run

  • Run parallel sections with isotype control antibody

  • Document lot-to-lot variations in antibody performance

  • Maintain a laboratory record of optimal conditions for each tissue type

How can I distinguish between specific and non-specific signals when using HIST1H3A (Ab-17) antibody?

Distinguishing specific from non-specific signals is critical for accurate data interpretation. For HIST1H3A (Ab-17) antibody:

  • Characteristic specific signal patterns:

    • Nuclear localization consistent with histone distribution

    • Intensity variations correlating with cell cycle phases

    • Reduced signal in mitotic chromosomes (where histone accessibility changes)

    • Reproducible staining patterns across technical replicates

  • Common non-specific signal patterns:

    • Cytoplasmic staining (histones are predominantly nuclear)

    • Uniform staining across all cell types regardless of biological context

    • Staining in negative control samples

    • Edge artifacts or staining of necrotic tissue regions

  • Validation approaches to confirm specificity:

    • Antibody titration: Specific signals typically show a dose-dependent relationship with antibody concentration while non-specific background increases linearly

    • Peptide competition: Pre-incubation with immunizing peptide should eliminate specific signals while leaving non-specific binding unaffected

    • Biological validation: Signal patterns should correlate with known biology (e.g., increased staining in proliferating cells)

    • Signal quantification: Plot signal-to-noise ratios across different antibody concentrations to identify optimal working conditions

  • Technical considerations for reducing non-specific signals:

    • Use freshly prepared buffers

    • Increase washing duration and frequency

    • Include carrier proteins (BSA, casein) in antibody diluent

    • Optimize blocking conditions based on tissue type

By implementing these approaches systematically, researchers can confidently distinguish between true HIST1H3A signals and experimental artifacts .

What controls should be included when using HIST1H3A (Ab-17) antibody in experimental workflows?

Proper controls are essential for valid and reproducible results with HIST1H3A (Ab-17) antibody. A comprehensive control strategy should include:

  • Antibody-specific controls:

    • Negative control: Omit primary antibody but include all other reagents

    • Isotype control: Use non-specific rabbit IgG at the same concentration

    • Peptide competition control: Pre-incubate antibody with excess immunizing peptide

    • Concentration gradient: Test multiple antibody dilutions to establish specificity threshold

  • Sample-specific controls:

    • Positive control tissues/cells: Human colon cancer tissue or HeLa cells known to express HIST1H3A

    • Low-expression controls: Differentiated, non-dividing cells with lower histone turnover

    • Fixation controls: Compare different fixation methods to rule out fixation artifacts

  • Biological validation controls:

    • siRNA knockdown: Reduced signal should correspond with HIST1H3A knockdown efficiency

    • Treatment controls: Cells treated with histone deacetylase inhibitors should show altered staining patterns

    • Cell cycle synchronization: Compare staining across different cell cycle phases

  • Technical controls:

    • Inter-assay controls: Include standard samples across multiple experiments

    • Batch controls: When processing multiple samples, distribute conditions across batches

    • Automated vs. manual processing comparison: Evaluate consistency between methods

  • Data analysis controls:

    • Blinded quantification: Have staining patterns scored by researchers unaware of sample identity

    • Multiple quantification methods: Compare different algorithms for signal quantification

    • Technical replicate variance analysis: Establish acceptable ranges for variation

Control TypePurposeImplementation
Technical negativeDetect non-specific bindingOmit primary antibody
Biological negativeValidate specificitysiRNA knockdown cells
Positive controlConfirm assay performanceHeLa cells or human colon cancer tissue
Peptide competitionVerify epitope specificityPre-incubate with R17 peptide
Cross-reactivityAssess off-target bindingTest on non-human samples

Implementing this comprehensive control strategy ensures reliable and interpretable results when using HIST1H3A (Ab-17) antibody .

How can HIST1H3A (Ab-17) antibody be used to study histone modifications in cancer progression?

HIST1H3A (Ab-17) antibody offers valuable opportunities for investigating the role of histone H3.1 and its modifications in cancer development and progression:

  • Differential expression analysis:

    • The antibody has been validated on human colon cancer tissue, showing nuclear localization patterns that can be compared between normal, pre-malignant, and malignant tissues

    • Changes in staining intensity can indicate altered histone dynamics or epigenetic reprogramming during carcinogenesis

  • Integration with tumor microenvironment studies:

    • Multiplex immunofluorescence combining HIST1H3A (Ab-17) with cancer stem cell markers and immune cell markers can reveal epigenetic heterogeneity within the tumor microenvironment

    • Co-localization analyses with hypoxia markers can investigate how oxygen tension affects histone modification patterns

  • Therapy response biomarker development:

    • Changes in R17 accessibility (as detected by this antibody) following treatment with epigenetic modifiers may predict therapy response

    • Serial biopsies during treatment can track dynamic epigenetic changes using quantitative immunohistochemistry

  • Methodological approach:

    • Tissue microarrays containing multiple patient samples at different disease stages

    • Recommended antibody dilution: 1:50 for IHC-P

    • Digital pathology quantification of nuclear staining intensity and heterogeneity

    • Correlation with patient outcome data for prognostic biomarker development

  • Correlation with other epigenetic marks:

    • HIST1H3A (Ab-17) can be used in sequential staining protocols with antibodies targeting other histone modifications, such as H3K9me3 and H3K27me3

    • Such studies can reveal the complex interplay between different histone marks in cancer epigenomes

This antibody provides researchers with a tool to investigate fundamental questions about the role of histone H3.1 dynamics in cancer evolution, potentially leading to new diagnostic or therapeutic approaches targeting epigenetic dysregulation .

What are the considerations for using HIST1H3A (Ab-17) antibody in conjunction with other histone modification antibodies?

Using HIST1H3A (Ab-17) antibody alongside other histone modification antibodies requires careful experimental design and interpretation:

  • Epitope accessibility considerations:

    • Modifications near R17 (such as H3K14ac, H3K18ac, H3K9me) may affect HIST1H3A (Ab-17) binding

    • When designing multi-antibody experiments, consider potential epitope masking effects

    • Sequential rather than simultaneous staining may be necessary for closely positioned epitopes

  • Compatible antibody combinations:

    Target 1Target 2CompatibilityNotes
    HIST1H3A (Ab-17)H3K9me3HighDifferent epitope regions
    HIST1H3A (Ab-17)H3K27me3HighDifferent epitope regions
    HIST1H3A (Ab-17)H3K14acLowAdjacent epitopes may interfere
    HIST1H3A (Ab-17)H3S10phMediumPhosphorylation may affect nearby epitope
  • Technical approach for multiplexed analysis:

    • Use different host species for primary antibodies when possible

    • For same-species antibodies, consider directly conjugated primaries or sequential tyramide signal amplification

    • Implement spectral unmixing for fluorophores with overlapping emission spectra

    • Include single-stain controls for each antibody to establish baseline signals

  • Data interpretation challenges:

    • Absence of HIST1H3A (Ab-17) signal may indicate either absence of H3.1 or presence of modifications that mask the R17 epitope

    • Quantitative correlation between signals requires normalization to total H3 levels

    • Cell cycle phase affects histone modification patterns and must be considered in interpretation

  • Validation strategies for multi-antibody experiments:

    • Western blot analysis with sequential probing for different modifications

    • ChIP-reChIP to confirm co-occurrence of modifications on the same histone molecules

    • Mass spectrometry to quantify combinations of modifications on individual histone tails

By thoughtfully addressing these considerations, researchers can leverage HIST1H3A (Ab-17) antibody in combination with other histone modification antibodies to gain comprehensive insights into chromatin regulation dynamics .

How can HIST1H3A (Ab-17) antibody contribute to understanding the interplay between histone modifications and gene expression?

HIST1H3A (Ab-17) antibody can be instrumental in elucidating the complex relationship between histone H3.1 dynamics, its modifications, and transcriptional regulation:

  • Integration with transcriptomic approaches:

    • Combine immunofluorescence using HIST1H3A (Ab-17) with RNA-FISH to correlate H3.1 distribution with active transcription sites

    • Implement cell sorting based on H3.1 levels (using the antibody for intracellular staining) followed by RNA-seq to identify genes differentially expressed in cell populations with varying H3.1 abundance

    • The recommended antibody dilution for immunofluorescence in such studies is 1:50-1:100

  • ChIP-seq applications:

    • While not explicitly validated for ChIP in the product information, the antibody's specificity for the R17 region suggests potential utility in chromatin immunoprecipitation

    • Sequential ChIP (ChIP-reChIP) with HIST1H3A (Ab-17) followed by modification-specific antibodies can identify genomic regions where specific modifications occur on H3.1 versus other H3 variants

    • Integration of such ChIP-seq data with RNA-seq can map correlations between H3.1 occupancy and transcriptional activity

  • Single-cell approaches:

    • Implement single-cell immunofluorescence to quantify cell-to-cell variation in H3.1 levels and correlate with transcriptional heterogeneity

    • Use imaging mass cytometry with HIST1H3A (Ab-17) antibody to analyze spatial distribution of H3.1 in relation to transcriptional regulators in tissue contexts

  • Functional studies methodology:

    • Combine HIST1H3A (Ab-17) staining with reporter gene assays to track how changes in H3.1 dynamics affect gene expression

    • Pre-treatment and post-treatment analysis in cells exposed to transcriptional inhibitors or activators can reveal dynamic relationships between H3.1 and gene expression

    • Time-course experiments during cell differentiation can elucidate how H3.1 patterns change during gene expression reprogramming

  • Epigenetic editing applications:

    • Use HIST1H3A (Ab-17) antibody to confirm targeted recruitment of H3.1 modifiers in CRISPR-dCas9 epigenetic editing experiments

    • Validate specificity of engineered histone modifiers by demonstrating altered HIST1H3A (Ab-17) binding patterns following editing

These approaches leverage the antibody's specificity for the R17 region of H3.1 to investigate how this histone variant and its modifications contribute to transcriptional regulation in normal development and disease states .

How might HIST1H3A (Ab-17) antibody be utilized in emerging single-cell epigenetic profiling technologies?

HIST1H3A (Ab-17) antibody holds significant potential for integration into emerging single-cell epigenetic technologies, providing insights into cell-to-cell variation in histone H3.1 dynamics:

  • Single-cell CUT&Tag applications:

    • The antibody can potentially be adapted for single-cell Cleavage Under Targets and Tagmentation (scCUT&Tag) protocols

    • This would allow genome-wide profiling of H3.1 distribution at single-cell resolution

    • Implementation requires optimization of antibody concentration (starting at 1:100 dilution) and incubation conditions for the specialized protocol

  • Mass cytometry integration:

    • Metal-conjugated HIST1H3A (Ab-17) antibody can be incorporated into CyTOF panels

    • This enables simultaneous quantification of H3.1 alongside dozens of other cellular proteins

    • Such analyses can reveal correlations between H3.1 patterns and cell fate determinants at single-cell resolution

  • Spatial epigenomics approaches:

    • Combining HIST1H3A (Ab-17) immunofluorescence with in situ sequencing technologies

    • This allows correlation of H3.1 distribution with spatial transcriptomics data

    • The antibody's validated performance in immunofluorescence (1:50-1:200 dilution) makes it suitable for such spatial analyses

  • Microfluidic-based single-cell epigenetic profiling:

    • Integration of HIST1H3A (Ab-17) antibody into microfluidic platforms for high-throughput single-cell histone profiling

    • Can be combined with barcoding strategies for multiplexed sample processing

    • Optimization of antibody dilution (starting at 1:100) for microfluidic environments is necessary

  • Live-cell imaging applications:

    • While the current antibody format is for fixed cells, research could explore adapting the HIST1H3A (Ab-17) epitope recognition principles for developing live-cell probes

    • This would enable real-time tracking of H3.1 dynamics during cellular processes

These emerging applications extend beyond traditional antibody uses, leveraging HIST1H3A (Ab-17)'s specificity for the R17 region to gain unprecedented insights into histone dynamics at single-cell resolution, with potential implications for developmental biology, cancer research, and regenerative medicine .

What role can HIST1H3A (Ab-17) antibody play in studying the relationship between histone modifications and 3D chromatin organization?

HIST1H3A (Ab-17) antibody offers unique opportunities to investigate how H3.1 distribution and modifications influence three-dimensional chromatin architecture:

  • Chromatin conformation capture integration:

    • Combine HIST1H3A (Ab-17) ChIP with Hi-C (ChIP-HiC) to identify genome regions where H3.1 contributes to specific chromatin interactions

    • This approach can reveal whether genomic regions enriched for H3.1 preferentially form topologically associating domains (TADs) or other 3D structures

    • Optimization of crosslinking conditions is critical when adapting the antibody for these applications

  • Super-resolution microscopy applications:

    • HIST1H3A (Ab-17) antibody can be used for super-resolution imaging techniques such as STORM or PALM

    • Recommended dilution for super-resolution: 1:50-1:100, with potential need for signal amplification systems

    • Such imaging can reveal nanoscale distribution of H3.1 in relation to chromatin domains and nuclear landmarks

    • Co-imaging with architectural proteins (CTCF, cohesin) can elucidate relationships between H3.1 and chromatin loop formation

  • Lamina-associated domain (LAD) studies:

    • Use HIST1H3A (Ab-17) in combination with lamin antibodies to investigate H3.1 enrichment at nuclear periphery

    • This can reveal connections between specific histone variants and repressive nuclear compartmentalization

    • Quantitative co-localization analysis workflows can measure association strength between H3.1 and nuclear lamina

  • Electron microscopy integration:

    • HIST1H3A (Ab-17) can be adapted for immunogold electron microscopy

    • This allows visualization of H3.1 distribution in relation to ultrastructural chromatin features

    • Gold particle size selection and optimization of embedding protocols are critical for success

  • Multi-modal chromatin analysis:

    • Combine microscopy using HIST1H3A (Ab-17) with genomic approaches for correlative analysis

    • This integrated approach can bridge the resolution gap between genomic and microscopic chromatin studies

    • Implementation requires standardized fixation protocols compatible with both imaging and genomic applications

These approaches exploit the antibody's specificity for the R17 region of H3.1 to investigate fundamental questions about how histone variants contribute to genome organization in three-dimensional nuclear space, with implications for understanding gene regulation mechanisms in development and disease .

What are the potential applications of HIST1H3A (Ab-17) antibody in studying age-related epigenetic changes?

HIST1H3A (Ab-17) antibody provides valuable opportunities for investigating age-associated epigenetic reprogramming through histone H3.1 dynamics:

  • Longitudinal aging studies:

    • HIST1H3A (Ab-17) can be applied to tissue samples collected across different age groups

    • Recommended dilution for immunohistochemistry on aged tissues: 1:20-1:100 (may require optimization due to increased autofluorescence in aged tissues)

    • Quantitative image analysis can track changes in H3.1 abundance and nuclear distribution patterns with advancing age

    • Correlation with markers of cellular senescence can reveal relationships between H3.1 dynamics and age-related cellular phenotypes

  • Investigation of histone turnover in aging:

    • Combine HIST1H3A (Ab-17) immunostaining with pulse-chase approaches to study age-related changes in H3.1 incorporation rates

    • This can reveal whether decreased histone turnover contributes to epigenetic drift during aging

    • Implementation requires careful experimental design with multiple timepoints and quantitative image analysis

  • Rejuvenation intervention studies:

    • Apply HIST1H3A (Ab-17) antibody to assess how interventions like caloric restriction, exercise, or pharmacological agents affect age-related H3.1 patterns

    • Such studies could identify epigenetic markers of biological versus chronological age

    • Before-after experimental designs with standardized staining protocols ensure valid comparisons

  • Tissue-specific aging comparisons:

    • Use the antibody to compare H3.1 dynamics across tissues with different aging rates

    • This can help identify whether differential histone management contributes to tissue-specific aging phenotypes

    • Implementation requires standardized sample processing to enable valid cross-tissue comparisons

  • Age-related disease applications:

    • Investigate whether neurodegenerative diseases, cancer, or cardiovascular disorders show altered H3.1 patterns

    • Compare age-matched healthy and disease samples to distinguish disease-specific from general aging changes

    • The antibody's validated performance in human tissues makes it suitable for translational aging research

This research direction leverages HIST1H3A (Ab-17) antibody's specificity to investigate fundamental questions about epigenetic stability and plasticity during aging, potentially identifying novel biomarkers of biological age and targets for interventions aimed at promoting healthy longevity .

What are the key considerations for interpreting results obtained with HIST1H3A (Ab-17) antibody?

  • Epitope specificity considerations:

    • The antibody recognizes the region around arginine 17 (R17) of human Histone H3.1

    • Modifications at or near R17 may affect antibody binding, potentially leading to underrepresentation of modified H3.1 populations

    • This specificity must be considered when interpreting absence of signal, as it may reflect epitope masking rather than absence of H3.1 protein

  • Technical variables affecting interpretation:

    • Fixation conditions significantly impact epitope accessibility

    • Antigen retrieval efficiency may vary between experiments and tissue types

    • Batch effects between staining runs should be normalized using standard samples

    • Signal intensity is not absolutely quantitative without careful calibration

  • Biological context for meaningful interpretation:

    • H3.1 levels naturally vary with cell cycle phase (higher in S phase)

    • Different cell types may have distinct baseline H3.1 levels

    • Cellular stress responses can alter histone dynamics

    • Developmental stage affects histone variant distribution

  • Quantification approaches and their limitations:

    • Semi-quantitative scoring (0, 1+, 2+, 3+) is suitable for broad pattern recognition

    • Digital image analysis provides more objective quantification but requires careful thresholding

    • Single-cell quantification reveals heterogeneity masked by population averages

    • Always report both the quantification method and its limitations

  • Corroborative evidence recommendations:

    • Support immunostaining results with orthogonal methods (Western blotting, mass spectrometry)

    • Validate unexpected findings using alternative antibodies targeting different H3.1 epitopes

    • Consider genetic approaches (overexpression, knockdown) to confirm specificity

By systematically addressing these considerations, researchers can extract reliable and meaningful biological insights from experiments utilizing HIST1H3A (Ab-17) antibody .

What best practices should researchers follow when reporting experimental results using HIST1H3A (Ab-17) antibody?

Transparent and comprehensive reporting of methods and results is essential for reproducibility in antibody-based research. When publishing studies using HIST1H3A (Ab-17) antibody, researchers should adhere to these best practices:

  • Detailed antibody information:

    • Report complete catalog information: HIST1H3A (Ab-17) Antibody, catalog number orb416534

    • Document antibody lot number, as performance may vary between lots

    • Specify antibody concentration used (not just dilution factor)

    • Include RRID (Research Resource Identifier) when available

  • Comprehensive methods reporting:

    • Provide complete sample preparation protocols, including fixation method, duration, and temperature

    • Detail antigen retrieval conditions (buffer composition, pH, duration, temperature)

    • Specify blocking conditions, antibody diluent composition, and incubation parameters

    • Document all washing steps (buffer composition, duration, number of washes)

    • Describe detection system completely, including secondary antibody details

  • Controls documentation:

    • Report all positive and negative controls used

    • Include images of control samples in supplementary materials

    • Document any validation experiments performed (peptide competition, knockdown)

    • Describe how non-specific binding was distinguished from specific signals

  • Quantification transparency:

    • Explain quantification methodology in detail (manual scoring, automated analysis)

    • Specify software used for image analysis, including version number

    • Document thresholding approaches and parameter settings

    • Report both raw and normalized data when appropriate

    • Include statistical methods for comparing staining between experimental groups

  • Results presentation standards:

    • Show representative images at multiple magnifications

    • Include scale bars on all images

    • Present images that reflect the full range of staining patterns observed

    • Avoid excessive contrast adjustment or selective field selection

    • Provide quantitative data with appropriate statistical analysis

Following these reporting practices ensures that experiments using HIST1H3A (Ab-17) antibody can be effectively evaluated and potentially reproduced by other researchers, advancing collective knowledge in the field .

What emerging research questions could be addressed using HIST1H3A (Ab-17) antibody in combination with new technologies?

HIST1H3A (Ab-17) antibody, when integrated with cutting-edge technologies, opens doors to addressing several frontier research questions in epigenetics and chromatin biology:

  • Single-cell heterogeneity in epigenetic states:

    • How does H3.1 distribution vary among seemingly identical cells within a tissue?

    • Does this heterogeneity correlate with cell fate decisions or disease progression?

    • Implementation approach: Combine HIST1H3A (Ab-17) antibody with single-cell sequencing technologies or high-content imaging platforms

  • Dynamic chromatin reorganization during cellular processes:

    • How does H3.1 incorporation change during DNA damage response and repair?

    • What is the temporal relationship between H3.1 deposition and gene activation/silencing?

    • Implementation approach: Live-cell imaging with H3.1 reporters, validated by fixed-cell analysis with HIST1H3A (Ab-17) antibody

  • Histone variant crosstalk in development and disease:

    • Do H3.1 and H3.3 variants show complementary or competitive genomic occupancy patterns?

    • How does this balance shift during cellular differentiation or malignant transformation?

    • Implementation approach: Sequential ChIP with variant-specific antibodies including HIST1H3A (Ab-17), followed by next-generation sequencing

  • Environmental influence on epigenetic landscapes:

    • How do environmental exposures reshape H3.1 distribution and modification patterns?

    • Can these changes be reversed through targeted interventions?

    • Implementation approach: Exposure studies with quantitative immunohistochemistry using HIST1H3A (Ab-17) antibody (1:20-1:200 dilution)

  • Spatial epigenomics across tissue architecture:

    • How does H3.1 distribution correlate with tissue microenvironments?

    • Are epigenetic territories maintained through specialized chromatin boundaries?

    • Implementation approach: Spatial transcriptomics combined with high-resolution imaging using HIST1H3A (Ab-17) antibody

  • Therapeutic manipulation of histone dynamics:

    • Can targeted approaches to modulate H3.1 incorporation provide therapeutic benefits?

    • How do existing epigenetic drugs affect H3.1 dynamics and distribution?

    • Implementation approach: Drug screening with HIST1H3A (Ab-17) antibody-based readouts

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.