H3F3A Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on your location and chosen shipping method. Please consult your local distributors for specific delivery times.
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
Histone H3.3 is a variant histone H3 that replaces conventional H3 in a wide range of nucleosomes within active genes. It is the predominant form of histone H3 in non-dividing cells and is incorporated into chromatin independently of DNA synthesis. Histone H3.3 is deposited at sites of nucleosomal displacement throughout transcribed genes, suggesting it represents an epigenetic imprint of transcriptionally active chromatin. Nucleosomes compact DNA into chromatin, limiting DNA accessibility to cellular machinery requiring DNA as a template. Thus, histones play a central role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Our findings indicate that the Clival GCT is genetically defined by somatic mutation in the H3F3A gene, connecting 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 observed the p.K27M mutation of H3F3A (H3.3K27M) in two tumors of young patients exhibiting 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 driver gene in giant cell tumor of bone. Notably, H3F3A mutations are absent in atypical giant cell tumor of bone. PMID: 28545165
  6. H3F3A mutational testing may be a useful tool to differentiate giant cell tumor of bone from giant cell-rich sarcoma. PMID: 28899740
  7. Our research indicates the presence of H3F3A/B mutations in cell tumors of bone, chondroblastomas, and aneurysmal bone cysts. PMID: 28882701
  8. Evidence suggests 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, loss of H3K9ac or SEC depletion appears to reduce gene expression. PMID: 28717009
  9. We investigated the incidence of H3.3 G34 mutations in primary malignant bone tumors through genotype analysis and H3.3 G34W immunostaining. PMID: 28505000
  10. The kinase activity of Aurora B on serine 31 of histone H3.3 was biochemically confirmed using nucleosomal substrates in vitro. PMID: 28137420
  11. This study demonstrated that heterozygous K27M mutations in H3F3A (n = 4) or HIST1H3B (n = 3) were present across all primary, contiguous, and metastatic tumor sites in all Diffuse intrinsic pontine gliomas. PMID: 26727948
  12. The 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. Our research found that 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, albeit less sensitive, diagnostic tool for differentiating GCTB and chondroblastoma from other giant cell-containing tumors. PMID: 26457357
  15. We describe three intriguing cases of pediatric glial and glioneuronal tumors harboring both BRAF V600E and H3F3A K27M mutations. PMID: 25389051
  16. Our observations further expand 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. Based on our findings, H3F3A p.Gly34 Trp or p.Gly34 Leu mutations are not a frequent occurrence 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 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. Notably, histone genes HIST1H3B (K27M) mutations are frequent in adult brainstem gliomas. PMID: 24242757
  24. The H3F3A K27M mutation is associated with thalamic gliomas. PMID: 24285547
  25. The results of this study indicate that H3F3A K27M mutant GBMs exhibit decreased H3K27me3, which may be of both diagnostic and biological relevance. PMID: 23414300
  26. H3F3A exon 2 mutation was 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 the H3.3 G34R mutated tumors presented primitive neuroectodermal tumors of the central nervous system and pediatric glioblastomas. PMID: 23354654
  31. Diffuse intrinsic pontine gliomas containing the 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. We propose a model where aberrant epigenetic silencing through H3K27M-mediated inhibition of PRC2 activity promotes gliomagenesis. PMID: 23539183
  32. A low frequency of H3.3 mutations was observed in myelodysplastic syndromes patients. PMID: 23660862
  33. Our data indicate that the H3.3K27M mutation reprograms the 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. The 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. Our research demonstrates that the two H3F3A mutations give rise to glioblastomas in separate anatomic compartments, with differential regulation of transcription factors OLIG1, OLIG2, and FOXG1, potentially reflecting different cellular origins. PMID: 23079654
  38. We report recurrent mutations in a regulatory histone in humans. Our data suggest that defects in chromatin architecture underlie pediatric and young adult GBM pathogenesis. PMID: 22286061
  39. We discuss the significance of H3.3 deposition as a salvage pathway to maintain chromatin integrity. PMID: 22195966
  40. Histone H3.3 is 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 the nucleosome remodeling and deacetylase (NuRD) repressor complex. PMID: 11850414
  43. We have analyzed histone posttranslational modifications on H3.1 and H3.3. PMID: 17052464
  44. Data reveal that TPA activates transcription of TBX2 by 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 why is it important in epigenetic research?

H3F3A is a gene that encodes the H3 histone family member 3A protein in humans, also known as Histone H3.3, H3.3A, or H3F3 . This non-centromeric histone variant plays a crucial role in both gene activation and silencing mechanisms . Unlike canonical histones, H3.3 is incorporated into chromatin independent of DNA replication, making it particularly important for dynamic chromatin regulation in non-dividing cells.

The methodology for studying H3F3A involves using specific antibodies that recognize either the total protein or its post-translationally modified forms. These antibodies enable researchers to track H3F3A incorporation patterns across the genome, correlate its presence with gene expression states, and understand its role in diverse cellular processes including development, differentiation, and disease progression.

How do I select the appropriate H3F3A antibody for my specific research application?

Selection of the appropriate H3F3A antibody depends on several methodological considerations:

  • Experimental application: Different applications require antibodies validated for specific techniques. For instance, Western blot applications may require different antibody characteristics than immunohistochemistry or chromatin immunoprecipitation (ChIP) .

  • Specificity requirements: Determine whether you need an antibody that recognizes:

    • Total H3F3A protein

    • H3F3A with specific post-translational modifications (e.g., phosphorylated at threonine 3)

    • Mutation-specific variants (e.g., K27M mutant)

  • Species cross-reactivity: Consider the species in which your experiments will be conducted. Many antibodies recognize human, mouse, and rat orthologs, but validation in your specific model system is essential .

  • Clonality considerations: Monoclonal antibodies offer high specificity for a single epitope, while polyclonal antibodies may provide stronger signals by recognizing multiple epitopes.

The most rigorous approach is to test multiple antibodies side-by-side using positive and negative controls relevant to your experimental system.

What are the standard applications for H3F3A antibodies in molecular biology?

H3F3A antibodies are employed in numerous molecular biology applications, each with specific methodological considerations:

ApplicationMethodology HighlightsCommon Optimizations
Western Blot (WB)Detects total or modified H3F3A in protein extractsRequires optimization of extraction methods to preserve histone modifications
Immunohistochemistry (IHC)Visualizes H3F3A distribution in tissue sectionsMay require antigen retrieval to expose epitopes in fixed tissues
Immunocytochemistry (ICC)Examines subcellular localization in cultured cellsOften combined with other markers for colocalization studies
Chromatin Immunoprecipitation (ChIP)Maps genomic locations of H3F3A bindingRequires careful crosslinking and sonication optimization
Flow Cytometry (FCM)Quantifies H3F3A levels in individual cellsOften used to correlate with cell cycle or differentiation markers
ELISAQuantitative measurement of H3F3A in solutionUseful for high-throughput screening applications

For optimal results, researchers should follow validated protocols specific to each application and perform preliminary titration experiments to determine the optimal antibody concentration .

How do post-translational modifications of H3F3A affect its function in chromatin regulation?

Post-translational modifications (PTMs) of H3F3A create a complex "histone code" that regulates chromatin structure and function. Specific antibodies recognizing these modifications enable researchers to investigate their biological significance.

Methodological approach to studying H3F3A PTMs:

  • Phosphorylation: Phosphorylation at threonine 3 (pThr3) alters the interaction between histones and DNA, affecting chromatin condensation during mitosis . Studying this modification requires phospho-specific antibodies like clone RM159, which recognizes H3F3A only when phosphorylated at threonine 3 .

  • Methylation: Lysine methylation states (mono-, di-, or tri-methylation) have distinct functional consequences. For example, H3K4me3 is associated with active transcription, while H3K27me3 correlates with gene silencing. Specific antibodies for these modifications enable ChIP-seq experiments to map their genomic distribution.

  • Acetylation: Lysine acetylation generally correlates with transcriptional activation. Antibodies targeting acetylated forms (e.g., H3K14ac) allow researchers to investigate regions of active chromatin.

When designing experiments to study these modifications, researchers should consider using inhibitors of histone-modifying enzymes as controls and validate antibody specificity using peptide competition assays or knockout/knockdown approaches.

What methodological considerations are critical when using H3F3A antibodies for ChIP-seq experiments?

ChIP-seq with H3F3A antibodies requires careful methodological planning:

  • Crosslinking optimization: The standard 1% formaldehyde fixation may not be optimal for all histone-DNA interactions. Titration experiments (0.5-2% formaldehyde) and time-course studies (5-20 minutes) should be performed to determine optimal crosslinking conditions.

  • Sonication parameters: Chromatin fragmentation to 200-500bp is critical for high-resolution mapping. Sonication conditions should be optimized for each cell type, and fragment size should be verified by agarose gel electrophoresis.

  • Antibody validation for ChIP: Not all antibodies that work for Western blot will work for ChIP. Validation using spike-in controls or known genomic regions enriched for H3F3A is essential.

  • Input normalization: Proper normalization to input chromatin is critical for accurate peak calling. Using spike-in standards can improve quantification across samples.

  • Bioinformatic analysis: H3F3A distribution analysis requires specialized approaches that account for the broad enrichment patterns typical of histones, unlike transcription factors which show sharp peaks.

When interpreting ChIP-seq data, researchers should be aware that many commercial H3F3A antibodies may also recognize the highly similar H3F3B protein, potentially complicating the interpretation of results unless highly specific antibodies are used.

How can mutation-specific H3F3A antibodies advance cancer research?

Mutation-specific H3F3A antibodies provide powerful tools for investigating the molecular mechanisms of cancers harboring specific histone mutations:

  • K27M mutation detection: Antibodies specifically recognizing the K27M mutant form of H3F3A (such as the D3B5T clone) enable precise identification of this mutation in pediatric gliomas and other cancers . This allows for accurate diagnosis and patient stratification in clinical research.

  • Mechanistic studies: These antibodies facilitate investigation of how K27M mutations alter the epigenetic landscape, including:

    • Immunoprecipitation of mutant-specific protein complexes

    • ChIP-seq to map genomic localization of mutant histones

    • Immunofluorescence to study nuclear distribution patterns

  • Therapeutic development: Mutation-specific antibodies enable high-throughput screening assays to identify compounds that specifically target cells bearing these mutations.

The methodological approach typically involves validating antibody specificity using positive controls (cell lines with confirmed mutations) and negative controls (wild-type cells), followed by optimization for the specific application (Western blot, IHC, or ChIP).

What controls should be included when validating H3F3A antibody specificity?

Rigorous validation of H3F3A antibody specificity requires incorporating multiple controls:

  • Positive controls:

    • Cell lines or tissues known to express H3F3A at high levels

    • Recombinant H3F3A protein for Western blot applications

    • Synthetic peptides containing the target epitope

  • Negative controls:

    • H3F3A knockout or knockdown samples where available

    • Isotype control antibodies to assess non-specific binding

    • Secondary antibody-only controls to evaluate background

  • Specificity controls:

    • Peptide competition assays where the antibody is pre-incubated with excess antigen

    • Cross-reactivity assessment with highly similar proteins (e.g., H3F3B)

    • Validation across multiple applications (Western blot, IHC, ChIP)

The most comprehensive validation approach combines multiple techniques to confirm that the observed signal is specifically due to H3F3A rather than related proteins or non-specific binding.

How should researchers optimize immunoprecipitation protocols for H3F3A?

Optimizing immunoprecipitation (IP) protocols for H3F3A requires addressing several critical parameters:

  • Lysis buffer composition:

    • Histones require specialized extraction methods due to their tight association with DNA

    • High-salt buffers (≥300mM NaCl) or acid extraction methods may be necessary

    • Include protease and phosphatase inhibitors to preserve post-translational modifications

  • Antibody amount optimization:

    • Titrate antibody concentration (typically 1-5μg per IP reaction)

    • Consider the binding capacity of the beads used for immunoprecipitation

  • Incubation conditions:

    • Optimize temperature (4°C is standard) and duration (4 hours to overnight)

    • Determine whether pre-clearing with protein A/G beads improves specificity

  • Washing stringency:

    • The number and composition of washes affect signal-to-noise ratio

    • More stringent washes reduce background but may decrease yield

  • Elution methods:

    • Competing peptides may enable gentler elution than boiling in SDS

    • For downstream mass spectrometry, consider on-bead digestion protocols

The optimal parameters should be determined empirically for each experimental system and antibody.

What factors influence the choice between polyclonal and monoclonal H3F3A antibodies?

The decision between polyclonal and monoclonal H3F3A antibodies depends on experimental goals and methodological considerations:

FactorPolyclonal AntibodiesMonoclonal Antibodies
Epitope recognitionRecognize multiple epitopes, potentially increasing signalTarget a single epitope, offering higher specificity
Batch-to-batch consistencyMay vary between lotsConsistent production from hybridomas
Application versatilityOften work across multiple applicationsMay be optimized for specific applications
Species cross-reactivityOften recognize conserved epitopes across speciesMay have more limited cross-reactivity
PTM specificityMay recognize multiple modification statesCan provide exquisite specificity for a single modification
Cost considerationsGenerally less expensiveTypically more costly to produce

For studies focusing on specific post-translational modifications, monoclonal antibodies often provide superior specificity. For applications requiring robust signal detection of total H3F3A, polyclonal antibodies may be advantageous. When possible, validating key findings with both polyclonal and monoclonal antibodies provides the strongest evidence .

How should researchers quantify H3F3A levels in immunoblotting experiments?

Accurate quantification of H3F3A in immunoblotting requires systematic methodology:

  • Loading control selection:

    • For whole cell lysates, standard loading controls like GAPDH or β-actin may be appropriate

    • For nuclear extracts, nuclear-specific controls such as Lamin B1 are preferable

    • For histone-specific comparisons, total H3 or H4 levels provide suitable normalization

  • Quantification methodology:

    • Use digital imaging systems with a linear dynamic range

    • Perform densitometry on non-saturated bands

    • Subtract local background signal for each band

  • Statistical analysis:

    • Run technical replicates (minimum of three) to assess measurement variability

    • Perform biological replicates to account for biological variation

    • Apply appropriate statistical tests (t-test, ANOVA) based on experimental design

  • Accounting for post-translational modifications:

    • When studying modified forms, normalize to total H3F3A rather than housekeeping proteins

    • Consider the possibility that modifications may affect antibody binding affinity

The most robust analysis combines quantitative immunoblotting with orthogonal methods such as mass spectrometry to confirm findings.

What approaches are recommended for analyzing H3F3A ChIP-seq data?

Analysis of H3F3A ChIP-seq data requires specialized approaches:

  • Peak calling considerations:

    • Histone ChIP-seq typically produces broad enrichment patterns rather than sharp peaks

    • Algorithms designed for broad peaks (e.g., SICER, MACS2 with broad peak settings) are preferable

    • Use appropriate false discovery rate thresholds (typically q < 0.05 or 0.01)

  • Normalization approaches:

    • Input normalization is essential for accurate comparison

    • Consider spike-in controls (e.g., Drosophila chromatin) for quantitative comparisons

    • Apply appropriate scaling factors to account for sequencing depth differences

  • Integrative analysis:

    • Correlate H3F3A enrichment with gene expression data

    • Compare distribution patterns with other histone modifications

    • Analyze enrichment at specific genomic features (promoters, enhancers, gene bodies)

  • Visualization strategies:

    • Generate average profile plots centered on transcription start sites

    • Create heatmaps showing enrichment patterns across different gene categories

    • Use genome browsers for locus-specific visualizations

The most informative analyses place H3F3A distribution in the context of other epigenetic marks and transcriptional activity.

How can researchers distinguish between signals from H3F3A and highly similar histone variants?

Distinguishing H3F3A from similar variants requires careful experimental design and analysis:

  • Antibody selection strategy:

    • Use antibodies raised against peptides containing amino acids that differ between variants

    • Validate specificity using recombinant proteins or peptides of each variant

    • Consider using epitope-tagged versions in experimental systems where possible

  • Mass spectrometry approaches:

    • Use targeted mass spectrometry to identify peptides unique to each variant

    • Develop MRM (multiple reaction monitoring) assays for specific quantification

    • Consider top-down proteomics to analyze intact histone proteins with their modifications

  • Genomic localization analysis:

    • Compare ChIP-seq profiles using antibodies specific to different variants

    • Analyze differential enrichment patterns at specific genomic features

    • Correlate variant-specific patterns with functional genomic data

  • Genetic approaches:

    • Use variant-specific knockdown/knockout to validate antibody specificity

    • Employ variant-specific rescue experiments to confirm functional differences

The most definitive approach combines immunological, genetic, and mass spectrometry methods to establish variant-specific identification.

What strategies can resolve weak or absent signals in H3F3A immunoblotting?

When encountering weak or absent signals in H3F3A immunoblotting, consider these methodological solutions:

  • Extraction optimization:

    • Standard lysis buffers may not efficiently extract histones

    • Implement acid extraction methods (e.g., 0.2N HCl) or high-salt extraction

    • Include histone deacetylase inhibitors (e.g., sodium butyrate) to preserve modifications

  • Transfer optimization:

    • Small proteins like histones (15.3 kDa) may transfer through the membrane

    • Use PVDF rather than nitrocellulose membranes

    • Decrease transfer voltage and increase transfer time

    • Consider using 0.2μm pore size membranes instead of standard 0.45μm

  • Blocking optimization:

    • Over-blocking can mask epitopes

    • Try different blocking agents (BSA vs. non-fat milk)

    • Reduce blocking time or concentration

  • Antibody optimization:

    • Increase antibody concentration or incubation time

    • Try different antibody clones targeting different epitopes

    • Consider using signal enhancement systems

  • Detection system sensitivity:

    • Switch to more sensitive detection methods (e.g., ECL Plus or femto-sensitivity substrates)

    • Increase exposure time within the linear range

    • Use digital imaging systems with adjustable sensitivity

Systematic testing of these parameters should identify the limiting factor in your experimental system.

How can researchers troubleshoot non-specific binding in H3F3A immunostaining?

Non-specific binding in H3F3A immunostaining can be addressed through these methodological refinements:

  • Fixation optimization:

    • Over-fixation can mask epitopes

    • Under-fixation can compromise morphology

    • Test different fixatives (paraformaldehyde, methanol) and durations

  • Antigen retrieval assessment:

    • Heat-induced epitope retrieval may be necessary

    • Compare citrate buffer (pH 6.0) vs. EDTA buffer (pH 9.0)

    • Optimize retrieval duration and temperature

  • Blocking improvements:

    • Include both protein blocking (BSA/serum) and permeabilization agents

    • Add additional blocking steps with species-specific serum

    • Consider specialized blocking reagents for highly autofluorescent tissues

  • Antibody dilution optimization:

    • Perform systematic titration series to identify optimal concentration

    • Extend primary antibody incubation time at lower concentrations

    • Test different diluents (PBS vs. TBS, with varying detergent concentrations)

  • Washing protocol enhancement:

    • Increase number and duration of washes

    • Include higher detergent concentrations in wash buffers

    • Use agitation during washing steps

Incorporating appropriate negative controls in parallel helps distinguish between specific signal and background.

What approaches can improve signal-to-noise ratio in H3F3A ChIP experiments?

Improving signal-to-noise ratio in H3F3A ChIP experiments requires systematic optimization:

  • Crosslinking refinement:

    • Optimize formaldehyde concentration (typically 0.5-1.5%)

    • Determine ideal crosslinking time (usually 5-20 minutes)

    • Consider dual crosslinking with additional agents like EGS for protein-protein interactions

  • Chromatin preparation optimization:

    • Adjust sonication conditions to achieve 200-500bp fragments

    • Filter lysates to remove insoluble material

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

  • Antibody selection and handling:

    • Use ChIP-validated antibodies (not all WB-validated antibodies work in ChIP)

    • Determine optimal antibody amount empirically (typically 2-5μg per reaction)

    • Consider pre-adsorbing antibodies to reduce non-specific binding

  • Washing stringency adjustment:

    • Increase salt concentration in wash buffers (up to 500mM NaCl)

    • Add non-ionic detergents to reduce hydrophobic interactions

    • Perform additional washing steps at critical points

  • PCR optimization for ChIP-qPCR:

    • Design primers for positive and negative control regions

    • Optimize primer concentration and annealing temperature

    • Verify primer efficiency using standard curves

The combination of these approaches typically yields substantial improvements in signal-to-noise ratio for H3F3A ChIP experiments.

How are emerging single-cell technologies being adapted for H3F3A research?

Single-cell technologies are revolutionizing H3F3A research through several methodological innovations:

  • Single-cell ChIP-seq approaches:

    • Microfluidic-based systems enable H3F3A profiling in individual cells

    • Droplet-based technologies allow for higher throughput

    • Computational methods for dealing with sparse data from single cells are being developed

  • CUT&Tag and CUT&RUN adaptations:

    • These techniques offer improved sensitivity for limited material

    • Can be performed on thousands of single cells in parallel

    • Provide higher resolution than traditional ChIP approaches

  • Single-cell multi-omics integration:

    • Simultaneous measurement of H3F3A distribution and transcription

    • Correlation of histone modifications with DNA methylation at single-cell resolution

    • Integration with chromatin accessibility data (scATAC-seq)

  • Live-cell imaging approaches:

    • FRAP (Fluorescence Recovery After Photobleaching) to study H3F3A dynamics

    • Single-molecule tracking to monitor H3F3A deposition and turnover

    • Optogenetic tools to manipulate H3F3A localization in real-time

These emerging technologies will provide unprecedented insights into the heterogeneity of H3F3A distribution and function across individual cells within complex tissues.

What computational approaches are being developed for integrating H3F3A data with other epigenomic features?

Advanced computational methods are enhancing integration of H3F3A data with broader epigenomic landscapes:

  • Machine learning algorithms:

    • Predictive models for H3F3A distribution based on DNA sequence and other histone marks

    • Classification of genomic regions based on combinatorial histone modification patterns

    • Deep learning approaches to identify complex relationships between H3F3A and gene regulation

  • Network analysis methods:

    • Construction of protein-protein interaction networks centered on H3F3A

    • Inference of regulatory relationships between H3F3A and transcription factors

    • Identification of epigenetic modules with coordinated behavior

  • Multi-modal data integration:

    • Methods for integrating ChIP-seq, RNA-seq, and chromatin accessibility data

    • Statistical approaches for correlating H3F3A with DNA methylation patterns

    • Visualization tools for exploring relationships across epigenomic dimensions

  • 4D Nucleome analysis:

    • Integration of H3F3A distribution with 3D chromatin organization data (Hi-C, ChIA-PET)

    • Temporal analysis of changes in H3F3A patterns during development or disease progression

    • Modeling the impact of H3F3A on chromatin folding and nuclear architecture

These computational advances will facilitate systems-level understanding of H3F3A function within the complex nuclear environment.

How might H3F3A antibodies contribute to developing epigenetic therapies?

H3F3A antibodies are poised to accelerate development of epigenetic therapies through multiple approaches:

  • Target identification and validation:

    • Identifying aberrant H3F3A modifications in disease states

    • Mapping genomic locations affected by pathological histone variants

    • Understanding the consequences of mutations like K27M on the epigenome

  • Drug discovery applications:

    • High-throughput screening assays using H3F3A antibodies to identify compounds that modulate specific modifications

    • Target engagement studies to confirm that drugs affect intended histone modifications in cells

    • Pharmacodynamic biomarker development for clinical trials

  • Therapeutic antibody development:

    • Engineered antibodies that can enter cells to target specific histone modifications

    • Antibody-drug conjugates directed against cells with particular H3F3A variants

    • CAR-T cell approaches targeting cancer cells with H3F3A mutations

  • Precision medicine approaches:

    • Patient stratification based on specific H3F3A modifications or mutations

    • Monitoring treatment response through changes in histone modification patterns

    • Developing combination therapies targeting multiple epigenetic mechanisms

These applications highlight the growing importance of H3F3A antibodies beyond basic research into translational medicine.

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.