H2AFX Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Mechanism and Biological Significance

The H2AFX gene encodes histone H2AX, a variant of the core histone H2A family. Phosphorylation at serine 139 (γ-H2AX) occurs rapidly after DNA damage, forming distinct nuclear foci that recruit repair proteins like MDC1 and BRCA1 . This modification is essential for checkpoint activation, cell cycle arrest, and efficient repair of DSBs. The recombinant monoclonal antibody specifically binds to this phosphorylated site, enabling precise detection of DNA damage in experimental models.

Production Process

H2AFX Recombinant Monoclonal Antibodies are synthesized via recombinant DNA technology:

  1. Cloning: H2AFX antibody genes are inserted into expression vectors.

  2. Expression: Host cells (e.g., mammalian or bacterial systems) produce the antibody.

  3. Purification: Affinity chromatography isolates the antibody for high specificity .

Key Features

FeatureDescriptionSource
Host SpeciesRabbit or mouse (varies by product)
ReactivityHuman, mouse, rat (validated in multiple species)
ApplicationsWestern blot, immunohistochemistry (IHC), immunofluorescence (IF), flow cytometry
SpecificityTargets phosphorylated serine 139 (γ-H2AX) or unphosphorylated H2AX

Primary Uses

ApplicationPurposeRecommended Dilution
Western Blot (WB)Detect γ-H2AX levels in lysates to quantify DNA damage1:1000–1:4000
Immunohistochemistry (IHC)Visualize γ-H2AX foci in tissue sections (e.g., tumor samples)1:50–1:200
Immunofluorescence (IF)Localize γ-H2AX foci in cultured cells or fixed tissues1:1000–1:4000
Flow Cytometry (FC)Measure γ-H2AX in single cells for population analysis1:50–1:200

Role in DNA Damage Response

  • γ-H2AX Foci Formation: The antibody detects rapid phosphorylation kinetics after ionizing radiation or chemotherapeutic agents, enabling real-time monitoring of DSBs .

  • Cancer Research: Overexpression of γ-H2AX correlates with genomic instability in tumors, making it a biomarker for radiosensitivity and chemotherapeutic efficacy .

Notable Studies

Study FocusKey FindingsSource
RadiosensitizationH2AFX antibodies demonstrated enhanced tumor ablation in hypoxic environments
Telomere Dysfunctionγ-H2AX signaling linked to YAP1 activation and tissue inflammation
PARthanatos PathwayKDM6B inhibition sustained γ-H2AX levels, promoting immunogenic cell death

Challenges and Considerations

  • Specificity: Cross-reactivity with non-phosphorylated H2AX or other histones requires careful validation .

  • Antibody Stability: Recombinant antibodies may require strict storage conditions (e.g., -20°C) .

  • Batch Consistency: Recombinant production minimizes lot-to-lot variability compared to traditional monoclonals .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The H2AFX recombinant monoclonal antibody is synthesized through a meticulous process involving recombinant DNA and in vitro cloning. The antibody genes are cloned into expression vectors, which are then introduced into host cells. This allows for the recombinant antibody's expression within a cell culture environment. Following expression, the antibody undergoes affinity chromatography purification. Thorough testing has validated its use in ELISA, IHC, and FC applications to detect human histone H2AX.

Histone H2AX and its phosphorylated form, γ-H2AX, are pivotal components of the cellular response to DNA damage. They serve as markers of DNA damage, facilitating the recruitment of repair proteins to damaged sites and contributing to the maintenance of genomic integrity and cell viability.

Form
Liquid
Lead Time
Typically, we are able to dispatch products within 1-3 working days after receiving your orders. Delivery time may vary depending on the purchasing method or location. For specific delivery timeframes, please consult your local distributors.
Synonyms
Histone H2AX (H2a/x) (Histone H2A.X), H2AFX, H2AX
Target Names
Uniprot No.

Target Background

Function
H2AFX is a variant histone H2A that replaces conventional H2A in a subset of nucleosomes. Nucleosomes are responsible for wrapping and compacting DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones thus play a crucial role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex system of post-translational modifications of histones, often referred to as the histone code, and nucleosome remodeling. H2AFX is essential for checkpoint-mediated arrest of cell cycle progression in response to low doses of ionizing radiation and for efficient repair of DNA double-strand breaks (DSBs) particularly when modified by C-terminal phosphorylation.
Gene References Into Functions
  1. ZNF506-dependent positive feedback loop regulates H2AX signaling after DNA damage. PMID: 30013081
  2. This study confirms that H2AFX variants are associated with an increased risk of BC. The above-reported sequence variants of MRE11 genes may not constitute a risk factor of breast cancer in the Polish population. PMID: 29678143
  3. gamma-irradiation also decreased the number of cells in the G1 phase, characterized by no interaction between H3S10ph and gammaH2AX. PMID: 30096372
  4. The topology of clusters of gammaH2AX foci can be categorized depending on the distance to heterochromatin. The newly presented method provides new possibilities to categorize spatial organization of point patterns by parameterization of topological similarity. PMID: 30072594
  5. this study suggests that individual and co-expression pattern of nuclear oxidized-PTP and gamma-H2AX might be used as a prognostic marker of gastric carcinoma PMID: 30126387
  6. Low pH2AX expression is associated with mouth Cancer. PMID: 30275188
  7. Results show that the H2AX 3'U TR is targeted by miR328 and its expression inhibited in osteosarcoma cells under radiation conditions. PMID: 29207178
  8. The results propose a model in which Aurora B-mediated H2AX-phosphorylated serine 121 probably provides a platform for Aurora B autoactivation circuitry at centromeres and thus plays a pivotal role in proper chromosome segregation. PMID: 27389782
  9. Data indicate that nuclear H2A histone family, member X protein (gammaH2AX) expression is positively associated with the programmed death-ligand 1 (PD-L1) expression in lung squamous cell carcinoma. PMID: 29275316
  10. phosphorylated histone H2AX was predictive of disease progression epithelial dysplasia of the oral cavity. PMID: 28543539
  11. Gamma-H2AX, phosphorylated KAP-1 and 53BP1 play an important role in the repair of heterochromatic radon-induced DNA double-strand breaks. PMID: 27922110
  12. in a series of human biopsies, non-metastatic SCCs displayed a higher degree of chromosomal alterations and higher expression of the S phase regulator Cyclin E and the DNA damage signal gammaH2AX than the less aggressive, non-squamous, basal cell carcinomas. However, metastatic Squamous cell carcinoma lost the gammaH2AX signal and Cyclin E, or accumulated cytoplasmic Cyclin E. PMID: 28661481
  13. We found that phosphorylation of histone H2AX on Ser139 (gamma-H2AX), a biomarker of DSBs, and phosphorylation of ATM at Ser1981, Chk2 at Thr68, and p53 at Ser15, part of signaling pathways associated with DSBs, are elevated in these cells. PMID: 28388353
  14. number of gammaH2AX foci did not significantly change following cardiac MR (median foci per cell pre-MR = 0.11, post-MR = 0.11, p = .90), but the number of 53BP1 foci significantly increased following MR PMID: 29309426
  15. Study provides evidence that phosphorylated H2AX binds to the promoter of miR-3196 and regulate its expression leading to lung cancer cell apoptosis. PMID: 27780918
  16. there may not be a link between low level phosphorylation gammaH2AX sites and double-strand DNA breaks in cells exposed to topoisomerase I or II inhibitors PMID: 27391338
  17. Residual gammaH2AX foci induced by low dose x-ray radiation in bone marrow mesenchymal stem cells do not cause accelerated senescence in the progeny of irradiated cells. PMID: 29165316
  18. miR-24-mediated knockdown of H2AX may be a novel negative regulator of mitochondrial function and insulin signaling. PMID: 28386126
  19. suggest that pH2AX alone or better in combination with MAP17 may become a novel and valuable prognostic biomarker for patients with laryngeal carcinoma treated with preservation approaches. PMID: 27166270
  20. The findings demonstrate that RNF168 couples PALB2-dependent homologous recombination to H2A ubiquitylation to promote DNA repair and preserve genome integrity. PMID: 28240985
  21. Data show that co-treated with vincristine and XL019, a inhibitor of JAK2 and P-glycoprotein (P-gp), up-regulated expression of p21 and phosphorylated H2A histone family, member X (pH2AX). PMID: 29187454
  22. The bile acid receptor TGR5, inducible nitric oxide synthase (iNOS) and gamma-histone family 2A variant (gamma-H2AX) are up-regulated. PMID: 27247425
  23. Co-localization of gammaH2AX and 53BP1 indicates promotion of (in)effective nonhomologous end-joining repair mechanisms at sites of DSB. Moreover, gammaH2AX/53BP1 foci distribution presumably reveals a non-random spatial organization of the genome in MDS and AML. PMID: 28359030
  24. Cyclin F-mediated degradation of SLBP limits H2A.X accumulation and apoptosis upon genotoxic stress in G2 cell cycle checkpoint. PMID: 27773672
  25. study demonstrates that the individual and combined expression patterns of the DDR molecules PARP1, gammaH2AX, BRCA1, and BRCA2 could be predictive of the prognosis of STS patients and suggests that controlling the activity of these DDR molecules could be employed in new therapeutic stratagems for the treatment of STS PMID: 27643881
  26. Further analysis suggested that H2AX, a PARP-1 protein interaction partner, was coordinated with PARP-1 in hepatocellular carcinoma tumorigenesis. Overall, some new characteristics of PARP-1 expression were noted in the Zhuang population. PARP-1 is a novel promising diagnostic marker for hepatocellular carcinoma in the Southern Chinese Zhuang population PMID: 28714367
  27. we found that gamma-H2AX foci at chromosome boundaries after carbon-ion irradiation contain DNA double strand breaks undergoing DNA-end resection, which promotes repair utilizing microhomology mediated end-joining during translocation. PMID: 27113385
  28. this study demonstrates an early DDR defect of attenuated gammaH2AX signals in G0/G1 phase HGPS cells and provides a plausible connection between H3K9me3 loss and DDR deficiency. PMID: 27907109
  29. Data indicate an important role for histone H2A.X (H2AX) Tyr39 phosphorylation in gamma-H2A.X formation and cancer progression. PMID: 27813335
  30. we suggest that the XAB2 complex mediates DNA damage response events important for the end resection step of homologous recombination , and speculate that its adjacent-localization relative to double-strand break marked by gH2AX is important for this function PMID: 27084940
  31. the epithelial-mesenchymal transition-related transcription factor Twist1 cooperates with Slug to regulate EMT upon H2A.X Loss. PMID: 27315462
  32. Upon DNA damage, an increase in the levels of chromatin bound motor protein nuclear myosin 1 (NM1) ensues, which appears to be functionally linked to Upsilon-H2AX signaling. PMID: 27365048
  33. TRAF6 and H2AX overexpression and gammaH2AX-mediated HIF1alpha enrichment in the nucleus of cancer cells lead to overactivation of HIF1alpha-driven tumorigenesis, glycolysis and metastasis. PMID: 27918549
  34. gammaH2AX, claimed to be a marker of DNA double-strand breaks, was found in cell extracts of HeLa cells at elevated temperature vs. 37.0 degrees C, and these gammaH2AX signals were intensified in the presence of 3-aminobenzamide, a PARP inhibitor. PMID: 27262441
  35. Data provide evidence that the acetylation of H2AX at Lys5 by TIP60 is required for the (ADPribosyl) ation activity and the dynamic binding of PARP-1 to chromatin after the induction of DNA damage. PMID: 26976643
  36. data cannot finally exclude H2AX methylation of SUV39H2 in cells, additional experimental evidence is required to validate this claim. PMID: 27177470
  37. This review outlines the role of gamma-H2AX in cell cycle, and its formation as a result of DNA damage. We investigate the role of gamma-H2AX formation in several cancer types and its correlation with other prognostic factors, and we try to find out whether it fulfills the requirements for its establishment as a classical cancer prognostic factor PMID: 28351323
  38. this study identified histone H2AX as an antigen of systemic lupus erythematosus by comparing highly ranked genes from all the built network-derived gene lists, which was confirmed the with real-world clinical samples PMID: 27226232
  39. dyserythropoiesis was increased in MDS patients with the deletion of chromosome 11q23, where H2AX is located. Although loss of H2AX did not affect the early stage of terminal erythropoiesis, enucleation was decreased. PMID: 26791933
  40. the formation of 53BP1, gammaH2AX foci and their co-localization induced by gamma-rays (2, 5, 10, 50, 200 cGy) in human lymphocytes, was analyzed. PMID: 26243567
  41. 5-Hydroxymethylcytosine (5hmC) accumulates at DNA damage foci and colocalizes with major DNA damage response proteins 53BP1 and gH2AX, revealing 5hmC as an epigenetic marker of DNA damage. PMID: 26854228
  42. Anacardic acid sensitizes prostate cancer cells to radiation therapy by repressing H2AX expression. PMID: 26884865
  43. Results reveal a pathway controlled by ATM, SIRT6, and SNF2H to block HUWE1, which stabilizes H2AX and induces its incorporation into chromatin only when cells are damaged. PMID: 26711340
  44. Gene expression analysis identified deregulation of histone H2A and H2B genes in all four cell lines ;histone pathways are associated with epirubicin resistance PMID: 26852132
  45. kinetics of the accumulation of selected DNA repair-related proteins is protein specific at locally induced DNA lesions, and that the formation of gH2AX- and NBS1-positive foci, but not 53BP1-positive NBs, is cell cycle dependent in HeLa cells PMID: 26482424
  46. The interaction of MDC1 with RNF8, but not with ATM requires WRAP53beta, suggesting that WRAP53beta facilitates the former interaction without altering phosphorylation of MDC1 by ATM. PMID: 26734725
  47. the interaction of 53BP1 with gammaH2AX is required for sustaining the 53BP1-dependent focal concentration of activated ATM that facilitates repair of DNA double-strand breaks in heterochromatin in G1. PMID: 26628370
  48. X-rays induce prolonged and ATM-independent persistence of gammaH2AX foci in human gingival mesenchymal stem cells PMID: 26314960
  49. Cell levels of gammaH2Ax define the G2 phase of the cell cycle. PMID: 26317799
  50. The study shows higher expression of gamma-H2AX and 53BP1 foci in rectal cancer patients compared with healthy individuals. Yet the data in vitro were not predictive in regard to the radiotherapy outcome. PMID: 26541290

Show More

Hide All

Database Links

HGNC: 4739

OMIM: 601772

KEGG: hsa:3014

STRING: 9606.ENSP00000364310

UniGene: Hs.477879

Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is H2AFX and why is it important in cellular biology?

H2AFX (also known as Histone H2AX or H2A.X) is a member of the histone H2A family, one of the four core histones involved in nucleosome formation and chromatin compaction. The critical importance of H2AFX lies in its role in DNA damage response pathways. When phosphorylated at serine 139 (creating γ-H2AX), it serves as a key marker of DNA double-strand breaks, enabling the recruitment of DNA repair proteins to damaged sites and contributing significantly to genomic integrity maintenance and cell viability . This phosphorylation occurs rapidly after DNA damage and creates a platform for the assembly of DNA repair complexes, making it an essential component in cellular mechanisms that protect against genomic instability.

How are H2AFX recombinant monoclonal antibodies produced?

H2AFX recombinant monoclonal antibodies are produced through a sophisticated process involving recombinant DNA technology and in vitro cloning. The process begins with cloning H2AFX antibody genes into expression vectors. These vectors are then introduced into host cells, creating a controlled environment for the recombinant antibody's expression within a cell culture system. Following expression, the antibody undergoes purification via affinity chromatography to ensure high specificity and consistency . Some H2AFX antibodies are generated using synthesized peptides derived from human H2AFX as the immunogen, resulting in highly specific non-conjugated rabbit IgG antibodies that are affinity-chromatography purified . This recombinant approach offers significant advantages over traditional monoclonal antibody production, including greater batch-to-batch consistency and reduced use of animals in production.

What are the primary applications of H2AFX recombinant monoclonal antibodies in research?

H2AFX recombinant monoclonal antibodies have diverse research applications, primarily in detecting human histone H2AX and its phosphorylated form. The antibodies are validated for multiple techniques:

  • ELISA (Enzyme-Linked Immunosorbent Assay): For quantitative detection of H2AFX in solution

  • IHC (Immunohistochemistry): For visualizing H2AFX in tissue sections, typically at dilutions of 1:50-1:200

  • FC (Flow Cytometry): For analyzing H2AFX expression in cell populations, also at recommended dilutions of 1:50-1:200

  • Western Blot: For detecting H2AFX in cell lysates and studying its modification states

  • Immunoprecipitation (IP): For isolating H2AFX and associated protein complexes

  • Immunofluorescence (IF): For subcellular localization studies and visualization of γ-H2AX foci formation

These applications enable researchers to investigate DNA damage response pathways, genotoxicity, cancer biology, and cellular responses to radiation and chemotherapeutic agents.

How do different phosphorylation sites on H2AFX affect antibody selection for specific research questions?

While phosphorylation at serine 139 (S139) of H2AFX is the most extensively studied modification, creating the γ-H2AX marker, researchers should consider that H2AFX contains multiple potential phosphorylation sites that may influence experimental outcomes and antibody selection. The phosphorylation at S139 (sometimes numbered as S140 depending on the reference sequence, as noted in entry NP_002096.1, Gene ID 3014 ) is critically important for DNA damage signaling.

When selecting an antibody for research:

  • For DNA damage studies: Antibodies specifically recognizing phospho-S139 (γ-H2AX), such as the recombinant monoclonal rabbit antibodies described in the search results, are optimal .

  • For total H2AFX studies: Non-phospho-specific antibodies that detect H2AFX regardless of modification state should be used.

  • For multi-modification studies: Consider that other modifications (e.g., ubiquitination, acetylation) may occur simultaneously with phosphorylation and potentially affect antibody binding.

Advanced research may require antibodies that can distinguish between different combinations of post-translational modifications on H2AFX, necessitating careful validation in the specific experimental system being studied.

What are the key considerations when using H2AFX antibodies for quantifying DNA damage across different cell types?

Quantifying DNA damage using H2AFX antibodies presents several important considerations that vary across cell types:

Baseline Expression Variability:
Different cell types exhibit varying baseline levels of H2AFX expression, which must be normalized appropriately when comparing DNA damage responses. Stem cells and rapidly dividing cancer cells typically show higher baseline H2AFX levels than terminally differentiated cells.

Kinetics of γ-H2AX Formation and Resolution:
The temporal dynamics of γ-H2AX formation and resolution after DNA damage differ significantly between cell types:

  • Lymphocytes: Typically show rapid γ-H2AX formation (minutes) and clearance (hours)

  • Neurons: Often exhibit prolonged γ-H2AX retention (days)

  • Cancer cells: May show aberrant γ-H2AX kinetics due to defective DNA repair pathways

Methodological Standardization:
For accurate quantification:

  • Use consistent fixation protocols (paraformaldehyde concentration and duration)

  • Standardize antibody dilutions (1:50-1:200 range is commonly effective for IHC and FC applications)

  • Implement appropriate permeabilization methods specific to the cell type

  • Include positive controls (e.g., irradiated cells) and negative controls

Data Interpretation:
When analyzing results, consider that γ-H2AX signal intensity may not linearly correlate with the number of DNA breaks across all cell types due to differences in chromatin accessibility and H2AFX density in the genome.

How can H2AFX antibodies be used to distinguish between different types of DNA damage responses?

H2AFX antibodies can be powerful tools for distinguishing between different types of DNA damage responses when used in sophisticated experimental designs:

Temporal Pattern Analysis:

  • Replication stress: Typically produces focal γ-H2AX patterns during S-phase

  • Double-strand breaks (DSBs): Generate distinct γ-H2AX foci at break sites

  • Apoptosis: Creates pan-nuclear γ-H2AX staining pattern

Co-localization Studies:
Combining γ-H2AX detection with other DNA damage response proteins reveals specific pathway activation:

  • 53BP1 + γ-H2AX: Non-homologous end joining (NHEJ) repair

  • RAD51 + γ-H2AX: Homologous recombination (HR) repair

  • BRCA1 + γ-H2AX: HR pathway activation

  • RPA + γ-H2AX: Replication stress and single-strand DNA exposure

Pulse-Chase Experiments:
Using time-course analysis with γ-H2AX antibodies can distinguish between:

  • Acute, high-dose damage (rapid formation and resolution)

  • Chronic, low-dose exposure (persistent low-level signal)

  • Defective repair (abnormal persistence of foci)

Multi-parameter Analysis:
Combining phospho-specific H2AFX antibodies with cell cycle markers (e.g., PCNA, Ki67) and other DNA damage markers enables comprehensive characterization of the damage response type within specific cell populations and cell cycle phases.

What are the optimal protocols for using H2AFX recombinant monoclonal antibodies in different applications?

Western Blot Protocol Optimization:

  • Sample preparation: Use 5% Milk-TBST for blocking and as antibody diluent

  • Primary antibody incubation: Overnight at 4°C

  • Secondary antibody selection: For cell lysates, use Goat anti-Rabbit IgG Heavy and Light Chain Antibody

  • For immunoprecipitates: Use Goat anti-Rabbit Light Chain HRP Conjugate with 5% Normal Pig Serum added to the blocking buffer

Immunohistochemistry (IHC) Optimization:

  • Recommended dilution range: 1:50-1:200

  • Antigen retrieval method: Heat-induced epitope retrieval in citrate buffer pH 6.0

  • Detection system: Polymer-based HRP detection systems are preferred for signal amplification

  • Counterstaining: Light hematoxylin to visualize nuclear localization

Flow Cytometry (FC) Protocol:

  • Cell fixation: 2-4% paraformaldehyde for 10-15 minutes

  • Permeabilization: 0.1-0.5% Triton X-100 or 70% ethanol (for improved nuclear access)

  • Antibody dilution: 1:50-1:200

  • Incubation time: 1 hour at room temperature or overnight at 4°C

  • Washing: PBS with 0.1% Tween-20, three times between steps

ELISA Protocol Considerations:

  • Coating concentration: 1-5 μg/ml of capture antibody

  • Blocking: 1-3% BSA in PBS

  • Sample preparation: Prepare cell or tissue lysates with appropriate extraction buffer

  • Detection: Use appropriate HRP-conjugated secondary antibody and substrate

What controls should be included when performing experiments with H2AFX antibodies?

Positive Controls:

  • For γ-H2AX antibodies: Include cells treated with ionizing radiation (2-10 Gy) or radiomimetic drugs (e.g., bleomycin, etoposide)

  • For total H2AFX antibodies: Use cell lines with confirmed H2AFX expression (most human cell lines express detectable levels)

  • Consider including known responsive cell lines such as lymphoblasts or fibroblasts

Negative Controls:

  • Primary antibody omission: Replace primary antibody with isotype-matched IgG (Rabbit IgG for most H2AFX recombinant monoclonal antibodies)

  • Blocking peptide: Pre-incubate antibody with the immunizing peptide to demonstrate specificity

  • H2AFX knockout/knockdown cells (if available): Demonstrate antibody specificity

Technical Controls:

  • Dilution series: Test multiple antibody dilutions (1:50, 1:100, 1:200) to determine optimal signal-to-noise ratio

  • Incubation time variants: Compare results with different incubation times

  • Storage impact: Test antibody performance after different storage conditions

Biological Response Controls:

  • Dose response: Include samples treated with different doses of DNA damaging agents

  • Time course: Collect samples at various time points after treatment to capture dynamics

  • Pharmacological inhibitors: Include ATM/ATR kinase inhibitors to block H2AFX phosphorylation

How can researchers optimize H2AFX antibody dilutions and incubation conditions for maximum sensitivity and specificity?

Optimizing H2AFX antibody protocols requires systematic testing of multiple parameters to achieve the ideal balance between sensitivity and specificity:

Antibody Dilution Optimization:

  • Start with the manufacturer's recommended range (typically 1:50-1:200 for IHC and FC)

  • Prepare a dilution series (e.g., 1:25, 1:50, 1:100, 1:200, 1:400)

  • Test each dilution on positive control samples

  • Evaluate signal-to-noise ratio at each dilution

Incubation Time and Temperature Matrix:

Temperature1 hour2 hoursOvernight
4°CMinimal signal, high specificityModerate signal, high specificityStrong signal, high specificity
Room TempModerate signal, good specificityStrong signal, moderate specificityVery strong signal, potential background
37°CStrong signal, moderate specificityVery strong signal, increased backgroundNot recommended

Buffer Composition Variations:

  • Test different blocking agents: BSA vs. normal serum vs. milk proteins

  • Adjust detergent concentration (0.05% to 0.3% Tween-20) to balance membrane permeabilization and antibody retention

  • Consider adding protein stabilizers (1-5% glycerol) for longer incubations

Antigen Retrieval Method Comparison:
For fixed tissues and cells, compare different antigen retrieval methods:

  • Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0)

  • HIER in EDTA buffer (pH 8.0-9.0)

  • Enzymatic retrieval with proteinase K

  • No retrieval (for some applications)

Document results systematically to create a reference protocol optimized for your specific experimental system and application.

What are common problems encountered with H2AFX antibodies and how can they be resolved?

High Background Signal:

  • Problem: Non-specific binding leading to diffuse background staining

  • Solutions:

    • Increase blocking time (2-3 hours or overnight)

    • Use alternative blocking agents (switch between BSA, normal serum, milk)

    • Increase washing steps (duration and number)

    • Further dilute primary antibody

    • Ensure proper storage conditions to prevent antibody degradation (-20°C or -80°C)

Weak or No Signal:

  • Problem: Insufficient antibody binding or detection

  • Solutions:

    • Verify antigen is not degraded in samples

    • Optimize antigen retrieval for IHC/ICC

    • Use more concentrated antibody solution (within 1:50-1:200 range)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Try more sensitive detection systems (e.g., tyramide signal amplification)

    • Confirm sample handling preserves phosphorylation (add phosphatase inhibitors)

Variable Results Between Experiments:

  • Problem: Inconsistent staining patterns or intensities

  • Solutions:

    • Standardize all protocol steps (timing, temperature, reagent sources)

    • Prepare larger volumes of diluted antibody to use across experiments

    • Include internal reference samples in each experiment

    • Monitor and control fixation times precisely

    • Avoid repeated freeze-thaw cycles of antibody

Cross-Reactivity Issues:

  • Problem: Antibody binds to unintended targets

  • Solutions:

    • Validate antibody specificity using knockout/knockdown controls

    • Perform peptide competition assays

    • Try alternative recombinant monoclonal antibodies targeting different epitopes

    • Increase washing stringency (higher salt or detergent concentration)

How should researchers analyze and interpret γ-H2AFX foci formation data from different experimental conditions?

Analysis and interpretation of γ-H2AFX foci require systematic approaches to generate reliable and reproducible results:

Quantification Methods:

  • Manual counting: Suitable for clear, distinct foci in limited samples

    • Count foci per nucleus (typically 30-100 nuclei per condition)

    • Establish consistent criteria for what constitutes a focus (size, intensity)

  • Automated image analysis:

    • Use specialized software (ImageJ with FIJI plugins, CellProfiler, etc.)

    • Set consistent parameters (threshold, minimum size, intensity)

    • Validate automated counts against manual counts for subset of images

Statistical Approaches:

  • For comparing treatments/conditions:

    • Use appropriate statistical tests based on data distribution

    • Consider both mean foci per nucleus and distribution patterns

    • Report both number of foci per cell and percentage of cells with foci

Temporal Analysis:

  • γ-H2AFX foci kinetics reveal important information about DNA damage processing:

    • Initial response (30 minutes - 1 hour): Formation rate indicates damage recognition efficiency

    • Peak response (1-4 hours): Maximum foci represent total damage burden

    • Resolution phase (4-24 hours): Disappearance rate indicates repair capacity

Combined Parameter Analysis:
When integrating γ-H2AFX data with other parameters, consider:

  • Cell cycle phase correlation (G1 vs. S vs. G2/M responses differ)

  • Co-localization with other repair factors (53BP1, RAD51, etc.)

  • Relationship between foci size/intensity and downstream outcomes (cell death, mutations)

Interpretation Guidelines:

  • Small, numerous foci often indicate acute DNA damage (e.g., radiation)

  • Large, persistent foci suggest complex or unrepairable damage

  • Pan-nuclear staining typically indicates replication stress or pre-apoptotic state

  • Foci number does not always linearly correlate with damage severity

What considerations are important when using H2AFX antibodies for multiplexed imaging with other DNA damage markers?

Multiplexed imaging combining H2AFX antibodies with other DNA damage markers provides rich contextual information but requires careful technical considerations:

Antibody Compatibility:

  • Species origin conflicts: When using multiple primary antibodies, they must come from different host species or be directly conjugated to different fluorophores

    • The H2AFX recombinant monoclonal antibodies described are rabbit-derived , requiring companion antibodies from mouse, rat, or directly conjugated antibodies

  • Cross-reactivity testing: Validate that secondary antibodies don't cross-react with inappropriate primaries

    • Test each antibody individually before combining them

Fluorophore Selection:

  • Choose fluorophores with minimal spectral overlap

  • Consider using Alexa Fluor 350-conjugated H2AFX antibodies as the blue channel component

  • Reserve brighter fluorophores (Alexa 488, Cy3) for markers with lower expression

  • Account for autofluorescence spectrum of your specific tissue/cell type

Sequential Staining Protocol:
When antibody conflicts cannot be avoided:

  • Apply first primary antibody and corresponding secondary

  • Fix the antibody-antigen complexes with 4% paraformaldehyde

  • Block with excess unconjugated Fab fragments

  • Apply subsequent primary-secondary pairs

  • Include extensive washing between steps

Image Acquisition Settings:

  • Capture single-stained controls to calculate and correct for spectral overlap

  • Maintain consistent microscope settings across samples for comparative analysis

  • Consider spectral unmixing for highly multiplexed imaging

Data Analysis Approaches:

  • Co-localization analysis:

    • Calculate Pearson's or Mander's coefficients between γ-H2AFX and other repair factors

    • Use distance-based measurements to quantify spatial relationships between different foci

  • Sequential recruitment analysis:

    • Design time-course experiments to determine ordering of factor recruitment

    • Analyze size-dependent co-localization to assess repair complex maturation

Controls for Multiplexed Imaging:

  • Single antibody controls to assess bleed-through

  • Fluorescence minus one (FMO) controls to set accurate thresholds

  • Competing unlabeled antibodies to confirm specificity in the multiplexed context

Recommended Dilutions and Applications for H2AFX Recombinant Monoclonal Antibodies

ApplicationRecommended DilutionIncubation TimeTemperatureDetection SystemComments
IHC1:50-1:200 1-2 hours or overnightRT or 4°CHRP-polymer or fluorescentHeat-mediated antigen retrieval recommended
FC1:50-1:200 30-60 minutes4°CFluorescent secondary or direct conjugatePermeabilization critical for nuclear antigen
WB1:500-1:2000Overnight4°CHRP-conjugated secondaryUse 5% milk-TBST for blocking
IP1:50-1:100Overnight4°CProtein A/G beadsUse light chain specific secondary for detection
IF1:100-1:5001-2 hoursRTFluorescent secondaryLonger primary incubation improves signal-to-noise ratio
ELISA1:1000-1:50001-2 hoursRTHRP/AP conjugated secondaryValidation with positive controls essential

Comparing γ-H2AFX Foci Formation in Response to Different DNA Damaging Agents

DNA Damaging AgentMechanismFoci PatternPeak Formation TimeResolution TimeSpecial Considerations
Ionizing RadiationDirect DSBsDistinct nuclear foci30 min - 1 hour6-24 hoursDose-dependent response, relatively uniform
UV RadiationPhotoproducts, Replication-dependent DSBsSmall foci, S-phase specific2-4 hours24-48 hoursCell cycle dependent, requires replication
EtoposideTopoisomerase II inhibitionMedium-sized foci1-2 hours12-24 hoursConcentration-dependent, cell type specific sensitivity
HydroxyureaReplication stressPan-nuclear or S-phase specific foci4-8 hours24-72 hoursPrimarily affects dividing cells
BleomycinRadiomimetic, oxidative damageDistinct large foci1-2 hours12-36 hoursGood positive control for γ-H2AFX antibody validation
CisplatinDNA crosslinksSmall, numerous foci6-12 hours48-72 hoursDelayed response compared to direct DSB inducers

How might emerging technologies enhance the use of H2AFX antibodies in research?

H2AFX antibody technology continues to evolve with several promising developments that may enhance their research utility. Super-resolution microscopy techniques like STORM and PALM are now enabling visualization of individual γ-H2AFX foci at nanometer resolution, potentially revealing substructures within repair complexes that were previously undetectable. Mass cytometry (CyTOF) approaches using metal-tagged H2AFX antibodies allow simultaneous detection of dozens of other DNA damage response proteins in single cells, providing unprecedented comprehensive pathway analysis.

Live-cell imaging compatible H2AFX antibody fragments and nanobodies are being developed to track DNA damage responses in real-time without fixation artifacts. Furthermore, proximity ligation assays using H2AFX antibodies paired with antibodies against other repair factors can provide quantitative measures of protein interactions at damage sites. These technological advances will likely transform our understanding of DNA damage response dynamics and their relationship to disease processes and treatment responses.

What are the most significant recent discoveries involving H2AFX antibodies in cancer and aging research?

Recent research using H2AFX antibodies has yielded significant insights into cancer biology and aging processes. Studies have revealed that certain cancer types exhibit "γ-H2AFX signatures" that correlate with treatment resistance and poor prognosis, offering potential biomarkers for personalized therapy approaches. In aging research, persistent γ-H2AFX foci have been identified as markers of senescence-associated DNA damage in multiple tissues, supporting the DNA damage theory of aging.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.