UNG Antibody is a polyclonal or monoclonal immunoglobulin raised against epitopes of the UNG enzyme. It facilitates the detection and quantification of UNG in biological samples, such as cell lysates or tissue sections, through techniques like Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF).
Key Features | Details |
---|---|
Target | Uracil-DNA glycosylase (UNG), isoforms UNG1 and UNG2 |
Primary Use | Research applications in DNA repair, immunology, and cancer biology |
Reactivity | Cross-species (human, mouse, rat, rabbit, zebrafish, etc.) |
Formats | Polyclonal (rabbit) or monoclonal (mouse) IgG |
UNG Antibody is validated for diverse research applications, as summarized below:
Example Protocol:
For Western blot, UNG Antibody is typically used at 2 µg/ml (Novus NB600-1031) or 1:2000 (Proteintech 12394-1-AP), followed by HRP-conjugated secondary antibodies and chemiluminescence detection .
Class Switch Recombination (CSR): UNG removes uracil residues introduced by activation-induced cytidine deaminase (AID) at immunoglobulin (Ig) loci, enabling CSR .
Somatic Hypermutation (SHM): UNG processes AID-induced uracil to generate transversions at G/C pairs, shaping antibody affinity .
Isoform Specificity: UNG2 (nuclear) and UNG1 (mitochondrial) exhibit distinct roles, but UNG1 can compensate for UNG2 loss in CSR .
Base-Excision Repair (BER): UNG repairs spontaneous uracil misincorporation, preventing mutations during replication .
Cancer Predisposition: UNG deficiency correlates with genomic instability and lymphoma risk, as observed in Ung−/− mice .
Phenotype: Patients with inactivating UNG mutations exhibit impaired CSR, leading to elevated IgM and recurrent infections .
Mechanism: UNG-deficient B cells fail to excise uracil at Ig loci, blocking switch recombination .
Lymphoma Risk: Ung−/− mice develop mature B-cell lymphomas due to unchecked AID activity and uracil accumulation .
Therapeutic Targeting: UNG inhibition may suppress oncogenic mutations in AID-expressing cancers .
Antibody | WB Sensitivity | IHC Specificity | IF Resolution |
---|---|---|---|
NB600-1031 | High (2 µg/ml) | Strong nuclear/mitochondrial staining | Poor (limited data) |
12394-1-AP | Moderate (1:500–1:2000) | Clear cytoplasmic/nuclear signals | Good (HeLa cells) |
NBP2-01194 | High (1:2000) | Validated in human tonsil | Moderate (COS7 cells) |
UNG (Uracil-DNA Glycosylase) excises genomic uracils created by activation-induced deaminase (AID) in B lymphocytes. This enzyme plays a dual role in underpinning antibody gene diversification and initiating faithful DNA repair . UNG is particularly important in germinal center B cells, where it protects cellular fitness coincident with AID-induced telomere damage that activates p53-dependent checkpoints . Research using UNG antibodies helps elucidate these mechanisms and provides insights into how UNG deficiency can lead to B-cell lymphoma development.
UNG antibodies are commonly utilized in several laboratory techniques including:
Western blotting for protein expression analysis
Immunohistochemistry (IHC) for tissue localization
Immunocytochemistry (ICC) for cellular localization
Immunofluorescence (IF) for subcellular localization
Flow cytometry for quantitative analysis
When selecting a UNG antibody, researchers should verify that it has been validated for their specific application, as antibody performance can vary significantly between different experimental contexts .
Validation of UNG antibodies should follow the "five pillars" consensus approach:
Genetic strategies: Use UNG knockout or knockdown models to confirm antibody specificity. The absence of signal in UNG-deficient samples provides strong evidence of specificity .
Orthogonal strategies: Compare antibody staining to protein/gene expression using an antibody-independent method (e.g., targeted mass spectroscopy) .
Independent antibody validation: Use multiple antibodies targeting different epitopes of UNG to confirm consistent staining patterns .
Expression of tagged proteins: Express tagged versions of UNG and confirm antibody co-localization with the tag .
Immunocapture followed by mass spectroscopy: Confirm that peptides captured by the antibody correspond to UNG protein sequences .
For robust validation, employ at least two of these approaches, ideally including genetic validation when possible.
When using UNG antibodies for western blotting:
Sample preparation: UNG has nuclear and mitochondrial isoforms (UNG2 and UNG1, respectively), with UNG2 being the predominant form in proliferating cells. Ensure proper subcellular fractionation if studying specific isoforms.
Molecular weight verification: Human UNG2 has a reported molecular weight of approximately 34.6 kDa , but post-translational modifications may alter the apparent size.
Controls: Include positive controls (e.g., cell lines known to express UNG) and negative controls (e.g., UNG-knockout cells if available).
Blocking and antibody concentration: Optimize blocking conditions and antibody dilutions to minimize background and maximize specific signal.
Detection method: Choose an appropriate detection method based on expected expression levels of UNG in your samples.
To study the UNG-RPA interaction, which is critical for guiding UNG to uracil in single-stranded DNA:
Co-immunoprecipitation:
Proximity ligation assay (PLA):
Use primary antibodies against UNG and RPA from different species.
Apply species-specific secondary antibodies with attached oligonucleotides.
If proteins are in close proximity, oligonucleotides can interact and be detected as fluorescent spots.
FRET or BiFC analysis:
Express fluorescently tagged UNG and RPA constructs.
Use antibodies against UNG and RPA as controls to validate the localization patterns of the tagged proteins.
When studying this interaction, note that RPA has been shown to be essential for UNG recruitment to uracil in single-stranded DNA, which is crucial for antibody class switch recombination .
UNG exists in two major isoforms (UNG1 and UNG2) that differ in their N-terminal sequences but share a common catalytic domain:
Isoform-specific epitopes:
Choose antibodies that target the unique N-terminal regions of UNG1 (mitochondrial) or UNG2 (nuclear).
Validate antibody specificity using overexpression systems with tagged isoform-specific constructs.
Cross-reactivity assessment:
Test antibodies in cell lines with known differential expression of UNG isoforms.
Use subcellular fractionation to separate nuclear (UNG2-enriched) and mitochondrial (UNG1-enriched) fractions.
Perform western blotting to confirm appropriate size differences between isoforms.
Immunofluorescence validation:
Co-stain with mitochondrial markers (for UNG1) or nuclear markers (for UNG2).
Analyze co-localization patterns to confirm proper isoform detection.
Genetic validation approaches:
Use isoform-specific siRNAs or CRISPR-based approaches to selectively deplete UNG1 or UNG2.
Confirm loss of specific bands or staining patterns with your antibody.
The AID-UNG interplay is crucial for antibody diversification and affects B-cell fitness. To investigate this relationship:
Co-localization studies in germinal center B cells:
Perform dual immunofluorescence with antibodies against UNG and AID.
Analyze co-localization at immunoglobulin loci using techniques like immuno-FISH.
Chromatin immunoprecipitation (ChIP):
Use UNG antibodies to perform ChIP and identify genomic regions where UNG is recruited.
Compare UNG recruitment patterns between wild-type and AID-deficient B cells.
Focus analysis on immunoglobulin switch regions and other known AID targets.
Functional assays in primary B cells:
Isolate germinal center B cells from immunized mice.
Use UNG antibodies to track UNG expression and localization during class switch recombination.
Compare results between wild-type and AID-deficient backgrounds.
Research has shown that UNG specifically protects the fitness of germinal center B cells that express AID, but not other B-cell subsets . This protection is likely related to UNG's role in repairing AID-induced DNA damage, particularly at telomeres.
To analyze UNG activity in clinical samples such as lymphoma specimens:
Immunohistochemistry panels:
Tissue microarray analysis:
Use validated UNG antibodies on tissue microarrays containing multiple patient samples.
Correlate UNG expression with clinical parameters and outcomes.
Consider dual staining with markers of DNA damage response.
Activity-based protein profiling:
Combined genomic and proteomic approaches:
Common pitfalls when using UNG antibodies include:
Non-specific binding:
Validate antibody specificity using genetic controls (UNG-knockout or knockdown).
Optimize blocking conditions and antibody dilutions.
Consider using monoclonal antibodies for higher specificity.
Application-specific variations:
Fixation and antigen retrieval issues:
For IHC/ICC, different fixation methods and antigen retrieval protocols can dramatically affect antibody performance.
Test multiple antigen retrieval methods (e.g., high/low pH buffers, different heating times).
Document optimal conditions in your protocols.
Cross-reactivity with related glycosylases:
UNG belongs to a family of uracil DNA glycosylases.
Verify antibody doesn't cross-react with other family members using recombinant proteins or knockout models.
To optimize immunoprecipitation with UNG antibodies:
Antibody selection and coupling:
Lysis conditions optimization:
Test different lysis buffers to maintain protein interactions while efficiently extracting UNG.
Consider nuclear extraction protocols for UNG2 (nuclear isoform).
Include phosphatase and protease inhibitors to preserve post-translational modifications.
Crosslinking considerations:
For transient interactions, consider using reversible crosslinkers prior to lysis.
Optimize crosslinking time and concentration to balance capture efficiency with specificity.
Validation of pulled-down complexes:
To distinguish specific from non-specific signals in IHC:
Multiple validation approaches:
Use at least two of the five validation pillars described earlier.
Compare staining patterns across multiple antibodies targeting different UNG epitopes.
Genetic controls:
Include UNG-knockout or knockdown tissues as negative controls.
Use tissues with known differential UNG expression (e.g., proliferating vs. quiescent tissues).
Absorption controls:
Pre-incubate antibody with recombinant UNG protein before staining.
Specific staining should be eliminated or significantly reduced.
Correlation with functional data:
Digital image analysis:
Use quantitative image analysis to set objective thresholds for positive staining.
Compare signal-to-background ratios across different tissues and controls.
UNG has complex roles in cancer, showing both tumor suppressor and tumor-enabling activities. To investigate these roles:
Expression analysis in cancer progression:
Use UNG antibodies to analyze expression patterns across cancer stages.
Create tissue microarrays with matched normal, premalignant, and malignant tissues.
Note that UNG expression is rarely lost in human B-cell lymphomas, with transcript levels correlating with proliferative characteristics of the neoplasm .
UNG localization in genome instability:
Use immunofluorescence co-staining with DNA damage markers.
Investigate UNG recruitment to sites of AID-induced damage in lymphomas.
Correlate UNG localization with mutation signatures.
Functional consequences of UNG variants:
Use antibodies to compare expression and localization of UNG variants found in cancers.
Investigate if cancer-associated UNG mutations affect RPA binding using co-immunoprecipitation approaches.
Therapeutic response biomarkers:
Evaluate whether UNG expression levels (detected by IHC) correlate with response to specific therapies.
Investigate UNG as a potential biomarker for DNA-damaging agent sensitivity.
Emerging approaches for studying UNG in the DNA damage response include:
Proximity-based labeling techniques:
Express UNG fused to enzymes like BioID or APEX2 that can biotinylate nearby proteins.
Use antibodies against UNG to validate the localization of fusion proteins.
Identify biotinylated proteins to map the UNG proximal interactome.
Super-resolution microscopy:
Use fluorescently labeled UNG antibodies with techniques like STORM or PALM.
Track UNG recruitment to DNA damage sites with nanometer precision.
Analyze co-localization with other DNA repair factors at super-resolution.
Live-cell imaging with complementary approaches:
Combine fluorescently tagged UNG with antibody validation in fixed cells.
Track UNG dynamics at sites of induced DNA damage.
Correlate with recruitment of RPA and other repair factors.
Chromatin proteomics:
Use UNG antibodies for ChIP-MS to identify proteins co-occupying UNG-bound genomic regions.
Focus analysis on immunoglobulin switch regions and other AID targets.
Compare proteomes between normal and neoplastic B cells.
When analyzing UNG expression across B-cell populations:
Standardized quantification methods:
For flow cytometry: Use isotype controls and fluorescence-minus-one (FMO) controls to set gates.
For IHC/IF: Use digital image analysis with consistent thresholding across samples.
For western blotting: Normalize UNG levels to appropriate loading controls.
Correlation with B-cell differentiation stages:
Functional interpretation:
Higher UNG expression in proliferating germinal center B cells reflects its role in managing AID-induced DNA damage.
In B-cell malignancies, UNG expression patterns correlate with the cell of origin and proliferative characteristics .
UNG expression is higher in activated B-cell-like DLBCL compared to GC B-cell-like DLBCL .
Subcellular localization analysis:
Nuclear UNG2 is the predominant form in proliferating cells.
Changes in nuclear/cytoplasmic distribution may indicate alterations in function or regulation.
When facing contradictory results across different assays:
Assess antibody validation quality for each assay:
Consider epitope accessibility differences:
Different fixation methods, sample preparations, or assay conditions can affect epitope accessibility.
Antibodies targeting different epitopes may give different results based on protein conformation or interactions.
Evaluate sample-specific factors:
Protein expression levels may be at the detection limit for some assays but not others.
Post-translational modifications may affect antibody binding differently across assays.
Protein interactions may mask epitopes in some contexts.
Resolve contradictions with orthogonal approaches:
Use non-antibody-based methods (e.g., mass spectrometry, RNA-seq, activity assays).
Apply genetic strategies (siRNA, CRISPR) to validate specificity.
Consider using tagged UNG constructs to track expression and localization.
Uracil-DNA glycosylase (UNG), also known as UDG, is a crucial enzyme involved in the base-excision repair (BER) pathway. Its primary function is to prevent mutagenesis by eliminating uracil from DNA molecules. This enzyme is essential for maintaining genomic stability and integrity.
Uracil can be erroneously incorporated into DNA through the deamination of cytosine or the misincorporation of dUMP residues. If not corrected, these uracil residues can lead to mutations during DNA replication. UNG excises uracil from DNA by cleaving the N-glycosidic bond, thereby initiating the BER pathway .
The enzyme operates through a “pinch-push-pull” mechanism, which involves five highly conserved motifs that collectively catalyze the glycosidic bond cleavage . These motifs include:
Mouse anti-human antibodies are commonly used in research to detect and study human proteins. The mouse monoclonal antibody against human UNG is an unconjugated antibody that has been validated for use in immunofluorescence (IF), immunohistochemistry (IHC), and western blotting (WB) . These antibodies are essential tools for studying the expression, localization, and function of UNG in various biological contexts.
The study of UNG and its interactions with other proteins is crucial for understanding the mechanisms of DNA repair and the prevention of mutagenesis. The mouse anti-human UNG antibody is particularly useful in research focused on cancer, as alterations in DNA repair pathways are often associated with tumorigenesis .