H3F3A (Ab-79) 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
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please contact your local distributors for specific delivery time information.
Synonyms
H3 histone family 3A antibody; H3 histone family 3B antibody; H3 histone; family 3B (H3.3B) antibody; H3.3 antibody; H3.3A antibody; H3.3B antibody; H33_HUMAN antibody; H3F3 antibody; H3F3A antibody; H3f3b antibody; Histone H3.3 antibody; Histone H3.3Q antibody; Histone H3.A antibody; Histone H3.B antibody; MGC87782 antibody; MGC87783 antibody
Target Names
H3F3A
Uniprot No.

Target Background

Function
Variant histone H3 replaces conventional H3 in a wide range of nucleosomes within active genes. It constitutes the predominant form of histone H3 in non-dividing cells and is incorporated into chromatin independently of DNA synthesis. H3.3 is deposited at sites of nucleosomal displacement throughout transcribed genes, suggesting it represents an epigenetic imprint of transcriptionally active chromatin. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machineries that require DNA as a template. Thus, histones play a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated via a complex set of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. We conclude that the Clival GCT is genetically defined by somatic mutation in the H3F3A gene, linking it to the GCT of long bones. PMID: 29609578
  2. Overexpression of H3F3A, encoding H3.3, is associated with lung cancer progression and promotes lung cancer cell migration by activating metastasis-related genes. PMID: 27694942
  3. We describe the presence of the mutation p.K27M of H3F3A (H3.3K27M) in two tumors of young patients with classical histopathology of ganglioglioma. PMID: 27219822
  4. H3F3 mutations are sensitive and specific markers of giant cell tumors of the bone and chondroblastomas. PMID: 28059095
  5. H3F3A is the most frequently mutated giant cell tumor of bone driver gene. H3F3A mutations are not present in atypical giant cell tumor of bone. PMID: 28545165
  6. H3F3A mutational testing may be a useful adjunct to differentiate giant cell tumor of bone from giant cell-rich sarcoma. PMID: 28899740
  7. Report H3F3A/B mutations in cell tumors of bone, chondroblastomas, and aneurysmal bone cysts. PMID: 28882701
  8. Data suggest that H3K9ac (histone 3 lysine 9 acetylation) serves as a substrate for direct binding of the SEC (super elongation complex) to chromatin; at select gene promoters, H3K9ac loss or SEC depletion appears to reduce gene expression. PMID: 28717009
  9. We determined the incidence of H3.3 G34 mutations in primary malignant bone tumors as assessed by genotype and H3.3 G34W immunostaining. PMID: 28505000
  10. The kinase activity of Aurora B on serine 31 of histone H3.3 was biochemically confirmed with nucleosomal substrates in vitro. PMID: 28137420
  11. This study showed that heterozygous K27M mutations in H3F3A (n = 4) or HIST1H3B (n = 3) across all primary, contiguous, and metastatic tumor sites in all Diffuse intrinsic pontine glioma. PMID: 26727948
  12. Study examined the relationship of K27M mutations in the distinct histone H3 variants (i.e. HIST1H3B and H3F3A) with specific pontine glioma biology. PMID: 26399631
  13. Study found spinal high-grade gliomas in children and adults frequently harbor H3F3A (K27M) mutations. PMID: 26231952
  14. H3F3A and H3F3B mutation analysis appears to be a highly specific, although less sensitive, diagnostic tool for the distinction of GCTB and chondroblastoma from other giant cell-containing tumors. PMID: 26457357
  15. We describe three interesting cases of paediatric glial and glioneuronal tumours harbouring both BRAF V600E and H3F3A K27M mutations. PMID: 25389051
  16. Our observations further extend the knowledge of H3F3A mutation and its location in pediatric glioblastomas. PMID: 25479829
  17. The CENP-A/histone H3.3 nucleosome forms an unexpectedly stable structure and allows the binding of the essential centromeric protein, CENP-C, which is ectopically mislocalized in the chromosomes of CENP-A overexpressing tumor cells. PMID: 25408271
  18. On the basis of our findings, H3F3A p.Gly34 Trp or p.Gly34 Leu mutations are not a frequent event in CGCL. PMID: 25442495
  19. These results suggest that immunohistochemical detection of H3.3 K27M is a sensitive and specific surrogate for the H3F3A K27M mutation and defines a prognostically poor subset of pediatric glioblastomas. PMID: 25200322
  20. This study identifies an H3.3K36me3-specific reader and a regulator of intron retention and reveals that BS69 connects histone H3.3K36me3 to regulated RNA splicing, providing significant, important insights into chromatin regulation of pre-mRNA processing. PMID: 25263594
  21. The mutually exclusive associations of HDAC1/p300, p300/histone, and HDAC1/histone on chromatin contribute to the dynamic regulation of histone acetylation. PMID: 24722339
  22. Loss of H3.3 from pericentromeric heterochromatin upon DAXX or PML depletion suggests that the targeting of H3.3 to PML-NBs is implicated in pericentromeric heterochromatin organization. PMID: 24200965
  23. These data suggest that adult brainstem gliomas differ from adult supratentorial gliomas. In particular, histone genes HIST1H3B (K27M) ) mutations are frequent in adult brainstem gliomas. PMID: 24242757
  24. H3F3A K27M mutation is associated with thalamic gliomas. PMID: 24285547
  25. The results of this study indicate that H3F3A K27M mutant GBMs show decreased H3K27me3 that may be of both diagnostic and biological relevance. PMID: 23414300
  26. H3F3A exon 2 mutation analyzed in solid tumors from 1351 South Korean patients. PMID: 23758177
  27. A remarkable picture of tumor type specificity for histone H3.3 driver alterations emerges, indicating that histone H3.3 residues, mutations and genes have distinct functions. PMID: 24162739
  28. Reduced H3K27me3 and/or DNA hypomethylation are the major driving forces of activated gene expression in K27M mutant pediatric high-grade gliomas. PMID: 24183680
  29. All reported H3.3 mutations identified in human tumors have been in the H3F3A gene leading to single codon changes within the N-terminal tail of the H3.3 protein. [Review] PMID: 24229707
  30. This study suggested that none of H3.3 G34R mutated tumors presented primitive neuroectodermal tumors of central nervous system and pediatric glioblastomas. PMID: 23354654
  31. Diffuse intrinsic pontine gliomas containing K27M mutation display lower overall amounts of H3 with trimethylated lysine 27(H3K27me3);H3K27M inhibits enzymatic activity of Polycomb repressive complex 2 through interaction with the EZH2 subunit; propose a model where aberrant epigenetic silencing through H3K27M-mediated inhibition of PRC2 activity promotes gliomagenesis. PMID: 23539183
  32. Low frequency of H3.3 mutations in myelodysplastic syndromes patients. PMID: 23660862
  33. Indicate that H3.3K27M mutation reprograms epigenetic landscape and gene expression, which may drive tumorigenesis. PMID: 23603901
  34. H3F3A K27M mutations occur exclusively in pediatric diffuse high-grade astrocytomas. PMID: 23429371
  35. Somatic mutation of H3F3A, a chromatin remodeling gene, is rare in acute leukemias and non-Hodgkin lymphoma. PMID: 23116151
  36. K27M mutation in H3.3 is universally associated with short survival in diffuse intrinsic pontine gliomas, while patients wild-type for H3.3 show improved survival. PMID: 22661320
  37. Demonstrate that the two H3F3A mutations give rise to glioblastomas in separate anatomic compartments, with differential regulation of transcription factors OLIG1, OLIG2, and FOXG1, possibly reflecting different cellular origins. PMID: 23079654
  38. Recurrent mutations in a regulatory histone in humans; data suggest that defects of the chromatin architecture underlie paediatric and young adult GBM pathogenesis. PMID: 22286061
  39. Discussion of the importance of H3.3 deposition as a salvage pathway to maintain chromatin integrity. PMID: 22195966
  40. Part of multiple H3.3-specific histone chaperone complexes. PMID: 21047901
  41. Studies indicate that H3.3 accomplishes a surprising variety of cellular and developmental processes. PMID: 20153629
  42. Histone H3 lysine 4 methylation disrupts binding of nucleosome remodeling and deacetylase (NuRD) repressor complex. PMID: 11850414
  43. Analysis of histone posttranslational modifications on H3.1 and H3.3. PMID: 17052464
  44. Data reveal that TPA activates transcription of TBX2 through activating MSK1, which leads to an increase in phosphorylated histone H3 and the recruitment of Sp1 to the TBX2 gene. PMID: 19633291

Show More

Hide All

Database Links

HGNC: 4764

OMIM: 137800

KEGG: hsa:3020

STRING: 9606.ENSP00000355778

UniGene: Hs.180877

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

Q&A

What is H3F3A and what does the H3F3A (Ab-79) Antibody recognize?

H3F3A is a gene that encodes the histone variant H3.3, a core component of nucleosomes that plays a central role in transcription regulation, DNA repair, replication, and chromosomal stability. The H3F3A (Ab-79) Antibody specifically recognizes the peptide sequence around the di-methyl-Lys (79) site derived from Human Histone H3.3 . This antibody is a polyclonal antibody raised in rabbits and demonstrates reactivity with human and rat samples . Unlike other H3 antibodies that recognize modifications at K79 such as trimethylation or acetylation, this antibody is specific to the dimethylated form of K79 on the H3.3 variant .

What are the recommended applications for the H3F3A (Ab-79) Antibody?

The H3F3A (Ab-79) Antibody has been validated for several experimental applications:

ApplicationRecommended DilutionNotes
Western Blot (WB)1:200-1:2000Detects H3F3A in cell lysates
ELISANot specifiedFor quantitative detection
ChIPNot specifiedFor studying protein-DNA interactions

For Western Blotting applications, researchers should optimize the dilution within the 1:200-1:2000 range depending on their sample type and detection method . When using this antibody for chromatin immunoprecipitation (ChIP), it's particularly valuable for investigating H3.3 deposition at specific genomic loci, especially intronic regions where H3.3 may act as a regulator of gene expression .

How should the H3F3A (Ab-79) Antibody be stored and handled?

For optimal performance and longevity of the antibody:

  • Store at -20°C or -80°C upon receipt

  • Avoid repeated freeze-thaw cycles that can degrade antibody quality

  • The antibody is supplied in liquid form containing 50% Glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as a preservative

  • Working dilutions should be prepared fresh before use and can be stored at 4°C for short periods (1-2 days)

Proper storage and handling are critical as degradation of antibody quality can lead to increased background signal, reduced specificity, or complete loss of immunoreactivity.

How does H3F3A differ from other histone H3 variants and their modifications?

The histone H3 family includes several variants with distinct functions:

Histone VariantEncoding GenesKey Characteristics
H3.3H3F3A, H3F3BReplication-independent, enriched at active genes
H3.1HIST1H3A-JReplication-dependent, incorporated during S phase
H3.2HIST2H3A-DReplication-dependent, canonical form

H3.3 (encoded by H3F3A) is particularly interesting as it can be incorporated into chromatin independently of DNA replication and is often associated with transcriptionally active regions . Lysine 79 modification on H3.3 (the target of this antibody) differs functionally from modifications at the same position on canonical H3 variants, with research suggesting it plays specific roles in gene activation during cellular processes like cancer progression .

How should I design a ChIP experiment using the H3F3A (Ab-79) Antibody?

When designing a Chromatin Immunoprecipitation (ChIP) experiment with the H3F3A (Ab-79) Antibody:

  • Sample preparation:

    • Cross-link protein-DNA complexes with 1% formaldehyde for 10 minutes at room temperature

    • Quench with 125mM glycine for 5 minutes

    • Isolate nuclei and sonicate chromatin to generate fragments of 200-500bp

  • Immunoprecipitation:

    • Use 5μg of H3F3A (Ab-79) antibody per ChIP reaction

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

    • Incubate overnight at 4°C with rotation

  • Washing and elution:

    • Perform stringent washes to remove non-specific binding

    • Elute protein-DNA complexes and reverse cross-links

  • Analysis:

    • Analyze by qPCR using primers specific to regions of interest

    • For genome-wide analysis, prepare libraries for ChIP-seq

Based on research findings, focus on intronic regions of genes as H3.3 has been shown to occupy intronic regions in cancer cells where it can modify chromatin status and directly activate transcription . For example, studies have demonstrated that H3.3 is deposited at specific intronic regions of genes like GPR87 in lung cancer cells .

What controls should I include when using this antibody for Western blots?

Proper controls are essential for accurate interpretation of Western blot results:

  • Positive control:

    • Cell lines known to express H3F3A (e.g., HeLa cells)

    • Acid-extracted histones from these cells ensure enrichment of histone proteins

  • Negative control:

    • Cell lines with H3F3A knockdown

    • Peptide competition assay using the immunizing peptide

  • Loading control:

    • Total histone H3 antibody to normalize loading

    • Other housekeeping proteins (though may not be ideal for histone studies)

  • Specificity controls:

    • Blots with peptides containing modified and unmodified K79 to verify specificity

    • Testing against other histone modifications (K4, K9, K27, etc.) to confirm absence of cross-reactivity

For Western blotting, the predicted molecular weight of histone H3.3 is approximately 15 kDa . Use SDS-PAGE gels with appropriate resolution for low molecular weight proteins (15-18% acrylamide) for optimal separation.

How can I optimize immunofluorescence staining using the H3F3A (Ab-79) Antibody?

For successful immunofluorescence staining:

  • Fixation:

    • Fix cells with 2.5-4% formaldehyde for 30 minutes at room temperature

    • For certain applications, methanol fixation may provide better epitope accessibility

  • Permeabilization:

    • Use 0.1-0.5% Triton X-100 in PBS for 5-10 minutes to allow antibody access to nuclear proteins

  • Blocking:

    • Block with PBS containing 1% BSA for 30-60 minutes

    • Include 0.1% Tween-20 to reduce background

  • Primary antibody:

    • Start with a 1:300 dilution of H3F3A (Ab-79) antibody

    • Incubate overnight at 4°C

  • Secondary antibody:

    • Use fluorophore-conjugated anti-rabbit antibody (Alexa 488, 568, or 647)

    • Include DAPI to counterstain nuclei

  • Controls:

    • Include a negative control omitting primary antibody

    • Perform peptide competition to confirm specificity

Similar protocols have been successful with other histone H3 antibodies as demonstrated in immunofluorescent analysis of human cells where clear nuclear localization was observed .

How can I use the H3F3A (Ab-79) Antibody to study the role of H3.3 in cancer progression?

Recent research has revealed important roles for H3F3A in cancer progression, particularly in lung cancer:

  • Expression analysis:

    • Compare H3F3A expression levels between tumor and normal tissues

    • Correlate expression with clinical parameters (stage, survival)

    • Perform immunohistochemistry on tissue microarrays

  • Functional studies:

    • Use ChIP with H3F3A (Ab-79) Antibody followed by sequencing to identify genome-wide binding sites

    • Focus on intronic regions of metastasis-related genes

    • Correlate H3.3 binding with gene expression changes

  • Mechanistic investigations:

    • Examine chromatin status at H3.3-bound regions using additional histone modification antibodies

    • Perform sequential ChIP (re-ChIP) to identify co-occurring modifications

    • Investigate interactions with chromatin remodeling complexes

Research has demonstrated that H3F3A overexpression promotes lung cancer cell migration by activating metastasis-related genes . H3.3 was found to globally activate gene expression through occupation of intronic regions, which showed characteristics of regulatory DNA elements . These findings suggest that monitoring H3F3A expression levels could serve as a prognostic marker for early-stage lung cancer .

What are the challenges in distinguishing H3F3A (K79) modifications from similar modifications on canonical H3?

Distinguishing between H3.3 and canonical H3 variants presents several technical challenges:

  • Sequence similarity:

    • H3.3 differs from H3.1/H3.2 by only 5/4 amino acids

    • The region around K79 is highly conserved between variants

  • Antibody specificity:

    • Validate specificity using dot blot analysis with different histone peptides

    • Perform peptide competition assays to confirm binding specificity

    • Test against various modified and unmodified peptides

  • Technical approaches:

    • Mass spectrometry can distinguish variants and modifications

    • Sequential immunoprecipitation with variant-specific then modification-specific antibodies

    • Genetic approaches (H3F3A knockdown/knockout) to validate signals

For dot blot validation, researchers can spot increasing amounts (0.2-100 pmol) of peptides containing the respective histone modifications onto membranes and probe with the antibody at 1/20000 dilution . This approach can effectively demonstrate antibody specificity against various histone modifications.

How can I investigate the relationship between H3F3A deposition and intronic regulation of gene expression?

The discovery that H3.3 occupies intronic regions to regulate gene expression opens new research directions:

  • Identify target genes:

    • Perform ChIP-seq with H3F3A (Ab-79) Antibody

    • Analyze intronic enrichment patterns

    • Correlate with gene expression data

  • Functional validation:

    • Design reporter constructs with and without identified intronic regions

    • Perform site-directed mutagenesis of putative regulatory elements

    • Use CRISPR-Cas9 to delete intronic regions of interest

  • Mechanism exploration:

    • Analyze chromatin accessibility at H3.3-bound intronic regions (ATAC-seq)

    • Investigate recruitment of transcriptional machinery

    • Examine long-range chromatin interactions (3C, Hi-C)

Research has shown that H3.3 is deposited at specific intronic regions of genes like GPR87, where it modifies chromatin status and directly activates transcription . This highlights the importance of intronic regions as regulatory elements in gene expression, particularly in contexts like cancer progression.

How should I interpret ChIP-seq data generated using the H3F3A (Ab-79) Antibody?

Proper interpretation of ChIP-seq data requires careful analysis:

  • Quality control metrics:

    • Signal-to-noise ratio: aim for >3

    • Library complexity: assess PCR duplicates

    • Peak distribution: evaluate genomic context

  • Peak analysis:

    • Examine distribution across genomic features (promoters, gene bodies, introns)

    • Look for enrichment at intronic regions based on known H3.3 binding patterns

    • Create aggregation plots around transcription start sites and gene bodies

  • Comparative analysis:

    • Compare with published H3.3 ChIP-seq datasets

    • Integrate with gene expression data

    • Correlate with other histone modifications

When analyzing data, pay special attention to intronic regions as research has shown H3.3 can bind to specific intronic regions where it modifies chromatin status and activates transcription . For example, studies demonstrated H3.3 deposition at a specific intronic region of GPR87 activated its transcription in lung cancer cells .

What are common pitfalls when using histone variant antibodies and how can I overcome them?

Several challenges are common when working with histone variant antibodies:

  • Cross-reactivity issues:

    • Solution: Perform dot blot analysis with modified and unmodified histone peptides

    • Test antibody against arrays of histone modifications

    • Include appropriate controls in each experiment

  • Low signal-to-noise ratio:

    • Solution: Optimize antibody concentration

    • Increase washing stringency

    • Use highly specific blocking agents

  • Batch-to-batch variability:

    • Solution: Validate each new antibody lot

    • Maintain reference samples for comparison

    • Document lot numbers used for each experiment

  • Non-specific binding:

    • Solution: Pre-clear lysates/chromatin

    • Include competitors (e.g., salmon sperm DNA for ChIP)

    • Perform peptide competition assays

  • Epitope masking:

    • Solution: Try different fixation methods

    • Test alternative epitope retrieval techniques

    • Consider native ChIP for certain applications

Careful validation through dot blot analysis with various peptides can help confirm specificity, as demonstrated in studies with other histone antibodies where peptides containing 0.2-100 pmol of respective histone modifications were spotted onto membranes for testing .

How can I integrate ChIP-seq data from H3F3A studies with other epigenomic datasets?

Integrative analysis provides deeper insights into H3.3 function:

  • Multi-omics integration approaches:

    • Combine H3F3A ChIP-seq with RNA-seq to correlate binding with expression

    • Integrate with ATAC-seq to assess chromatin accessibility at binding sites

    • Include DNA methylation data to examine epigenetic crosstalk

  • Analytical tools:

    • Use tools like HOMER, ChIPseeker, or GREAT for genomic annotation

    • Apply integrative platforms like Seqmonk or Galaxy

    • Consider machine learning approaches for pattern recognition

  • Visualization strategies:

    • Create browser tracks showing H3F3A binding alongside other datasets

    • Generate heatmaps clustered by binding patterns

    • Produce metaplots centered on features of interest

  • Biological interpretation:

    • Focus on cell-type specific patterns

    • Examine developmental trajectories

    • Look for disease-associated signatures

Research demonstrates that integrating H3F3A binding data with expression profiles can reveal regulatory relationships, such as how H3.3 deposition at intronic regions correlates with activation of genes like GPR87 in lung cancer . Such integration helped researchers identify H3F3A and GPR87 expression as prognostic markers for early-stage lung cancer .

What are emerging applications for studying H3F3A in single-cell epigenomics?

Single-cell approaches are revolutionizing our understanding of epigenetic heterogeneity:

  • Technical advances:

    • Single-cell ChIP-seq adaptations for H3F3A

    • CUT&Tag approaches for improved sensitivity

    • Combined single-cell transcriptome and epigenome profiling

  • Biological applications:

    • Studying H3F3A dynamics during cellular differentiation

    • Investigating heterogeneity within tumor microenvironments

    • Mapping cell-state transitions mediated by H3.3

  • Analytical frameworks:

    • Trajectory inference from single-cell epigenomic data

    • Integration with spatial transcriptomics

    • Pseudotime analysis of H3F3A deposition patterns

These approaches would be particularly valuable for studying heterogeneity in cancer progression, as H3F3A has been implicated in cancer cell migration and metastasis-related gene activation . Single-cell approaches could reveal how subpopulations of cells with distinct H3.3 patterns contribute to tumor progression.

How might the study of H3F3A inform therapeutic strategies for cancer treatment?

H3F3A biology suggests several potential therapeutic approaches:

  • Target identification:

    • H3F3A-regulated genes as drug targets

    • Proteins that facilitate H3.3 deposition

    • Enzymes that modify H3.3 at K79

  • Therapeutic strategies:

    • Small molecule inhibitors of H3.3 chaperones

    • Epigenetic drugs targeting H3.3-associated complexes

    • Antisense oligonucleotides targeting H3F3A

  • Biomarker applications:

    • H3F3A expression as prognostic indicator

    • Combined H3F3A/GPR87 expression signature

    • H3.3K79 modification status as predictive biomarker

Research has shown that H3F3A and GPR87 expression levels, either alone or in combination, serve as robust prognostic markers for early-stage lung cancer . This suggests potential for developing treatments involving GPR87 antagonists, highlighting how understanding H3F3A biology can lead to novel therapeutic strategies .

What are the implications of H3F3A intronic regulation for understanding gene expression mechanisms?

The discovery of H3.3's role in intronic regulation represents a paradigm shift:

  • Conceptual advances:

    • Introns as active regulatory elements rather than passive spacers

    • Histone variant deposition as a mechanism for intronic regulation

    • Integration of chromatin structure with RNA processing

  • Methodological approaches:

    • CRISPR screens targeting intronic H3.3 binding sites

    • Massively parallel reporter assays for intronic elements

    • Long-read sequencing to link chromatin states with splicing patterns

  • Broader implications:

    • Potential for widespread intronic regulation across the genome

    • Implications for evolution of gene regulation

    • New layers of complexity in disease-associated gene dysregulation

Research has demonstrated that H3.3 globally activates gene expression through occupation of intronic regions in lung cancer cells, where H3.3 binding regions show characteristics of regulatory DNA elements . This suggests intronic regulation by H3F3A may be a target for developing novel therapeutic strategies in cancer and other diseases .

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.