DGUOK is overexpressed in lung adenocarcinoma and correlates with poor prognosis . Research using DGUOK antibodies has uncovered:
Mechanism: DGUOK regulates NAD+ biogenesis via NMNAT2, promoting tumor progression and metastasis .
Therapeutic Targeting: Inhibitors like didanosine (DDI) reduce mtDNA levels and tumor growth, suggesting DGUOK as a potential therapeutic target .
Mutations in DGUOK cause mitochondrial DNA depletion syndrome (MDS), characterized by mtDNA loss in liver, brain, and muscle . Antibodies are used to:
The Sigma-Aldrich HPA034766 antibody demonstrates robust specificity in IHC, validated across 44 normal tissues and 20 cancer types via the Human Protein Atlas .
| Feature | PA5-63419 | AMS.AP7086A-ev | HPA034766 | H00001716-M02 |
|---|---|---|---|---|
| Host | Rabbit | Rabbit | Rabbit | Mouse (3E9) |
| Reactivity | Human, Mouse, Rat | Human, Mouse | Human | Human |
| Applications | WB, IHC | WB | WB, IHC, IF | WB (transfected lysate) |
| Immunogen | Peptide | Recombinant protein | Recombinant protein | Peptide |
DGUOK (deoxyguanosine kinase) is a mitochondrial enzyme that plays a critical role in the phosphorylation of purine deoxyribonucleosides in the mitochondrial matrix. This 277 amino acid protein is essential for maintaining mitochondrial DNA integrity by catalyzing the conversion of deoxyguanosine to deoxyguanosine monophosphate using ATP. DGUOK has gained significant research interest due to its involvement in NAD+ biogenesis, mitochondrial oxidative phosphorylation, and its overexpression in certain cancers, particularly lung adenocarcinoma . Additionally, mutations in DGUOK can lead to mitochondrial DNA depletion syndrome, making it an important target for both cancer and genetic disorder research .
DGUOK antibodies can be utilized across multiple research applications including:
Western blotting (WB) for protein quantification and molecular weight confirmation
Immunoprecipitation (IP) for protein-protein interaction studies
Immunofluorescence (IF) for cellular localization studies
Enzyme-linked immunosorbent assay (ELISA) for protein quantification
Immunohistochemistry (IHC) for tissue localization and expression analysis
The specific application suitability depends on the antibody clone and format, with some antibodies optimized for certain techniques over others . For instance, the H-3 mouse monoclonal antibody has been validated for WB, IP, IF, and ELISA applications with human, mouse, and rat samples .
Determining optimal antibody dilution requires consideration of several factors:
Application-specific recommendations: Starting dilutions typically range from 1:500-1:2000 for WB and 1:50-1:500 for IHC
Signal-to-noise optimization: Begin with the manufacturer's recommended dilution and adjust based on initial results
Sample type considerations: Different tissue/cell types may require different dilutions due to varying expression levels
Detection method sensitivity: More sensitive detection systems (e.g., chemiluminescence) may allow for higher dilutions
Always perform a dilution series in preliminary experiments, and note that recombinant DGUOK may require different dilutions than endogenous protein detection .
A robust validation strategy for DGUOK manipulation should include multiple approaches:
mRNA level validation:
RT-qPCR targeting multiple regions of DGUOK transcript
RNA-seq analysis for genome-wide effects
Protein level validation:
Functional validation:
For genetic manipulation, researchers have successfully used CRISPR-Cas9 with the sgRNA sequence 5'-CCCCGAAGGCTCTCCATCGA-3' for DGUOK knockout, and shRNA approaches for knockdown studies .
When investigating this regulatory axis, include these critical controls:
Expression controls:
Genetic manipulation controls:
Metabolic controls:
Specificity controls:
This comprehensive control strategy helps distinguish direct DGUOK effects from secondary metabolic consequences .
Multiple DGUOK bands in Western blot may reflect several biological and technical factors:
Multiple isoforms:
Post-translational modifications:
Phosphorylation or other modifications can alter migration patterns
Mitochondrial targeting sequence cleavage affects apparent molecular weight
Technical considerations:
Sample preparation conditions (reducing vs. non-reducing) affect protein conformation
Gel percentage affects resolution of closely migrating isoforms
Cross-reactivity with related proteins
To distinguish between these possibilities, consider using:
Isoform-specific antibodies if available
Denaturing vs. native conditions
DGUOK-knockout cells as negative controls
Phosphatase treatment to eliminate modification-based mobility shifts
Inconsistent DGUOK staining can be addressed through systematic optimization:
Antigen retrieval optimization:
Fixation considerations:
Overfixation can mask epitopes; standardize fixation protocols
Fresh tissue vs. FFPE samples may require different protocols
Antibody specificity verification:
Detection system optimization:
Try different secondary antibody systems (polymer-based vs. avidin-biotin)
Adjust counterstaining to improve contrast for mitochondrial patterns
Cross-validation:
DGUOK antibodies enable several approaches to investigate mitochondrial metabolism in cancer:
Expression profiling:
Mechanistic studies:
Therapeutic target validation:
Cancer stem cell research:
These applications have revealed that DGUOK overexpression promotes lung adenocarcinoma progression through enhanced mitochondrial oxidative phosphorylation and NAD+ biogenesis, suggesting potential therapeutic targets .
To address contradictory findings about DGUOK function across tissues, consider these methodological approaches:
Tissue-specific expression analysis:
Quantitative comparison of DGUOK expression across tissues using validated antibodies
Correlation with tissue-specific metabolic profiles and mitochondrial content
Conditional knockout models:
Multi-omics integration:
Combine proteomics, metabolomics, and transcriptomics data from DGUOK-manipulated models
Pathway analysis to identify tissue-specific dependencies on DGUOK function
Context-dependent interaction mapping:
Compare DGUOK interactomes across tissue types using co-immunoprecipitation
Identify tissue-specific regulatory proteins that modify DGUOK function
Metabolic flux analysis:
Trace isotope-labeled nucleosides to quantify tissue-specific differences in DGUOK-dependent pathways
Measure NAD+ synthesis rates in different contexts
These approaches can help reconcile the seemingly contradictory observations that DGUOK knockout severely affects lung cancer cells but has more modest effects in other cell types like fibroblasts .
Optimizing detection of mitochondrial DGUOK requires specific fixation and permeabilization approaches:
Fixation options:
4% paraformaldehyde (10-15 minutes at room temperature) preserves mitochondrial morphology
Methanol fixation (10 minutes at -20°C) may provide better epitope accessibility
Avoid extended fixation times which can mask mitochondrial epitopes
Permeabilization considerations:
0.1-0.2% Triton X-100 (5-10 minutes) for balanced permeabilization
0.1% saponin for gentler permeabilization that better preserves mitochondrial membranes
Digitonin (10-50 μg/ml) for selective permeabilization of plasma membrane while preserving mitochondrial membranes
Blocking optimization:
Use 5% normal serum from the same species as the secondary antibody
Include 0.1% BSA to reduce non-specific binding
Consider adding 0.1% Tween-20 to reduce background
Co-localization markers:
Include mitochondrial markers (MitoTracker, TOM20, or COXIV) for co-localization confirmation
Nuclear counterstain (DAPI) to provide cellular context
Antibody validation:
These optimizations help visualize the authentic mitochondrial distribution of DGUOK while minimizing artifacts.
To quantitatively evaluate DGUOK's effect on mitochondrial function, implement these approaches:
mtDNA integrity and abundance:
Respiratory complex activity:
Western blot analysis of respiratory complex subunits (mt-ND1, NDUFB8, NDUFB10, mt-CO2)
Complex I enzyme activity assays (NADH:ubiquinone oxidoreductase activity)
Oxygen consumption rate (OCR) measurements using Seahorse XF analyzer
NAD+ metabolism assessment:
dNTP pool analysis:
Functional consequences:
Mitochondrial membrane potential using JC-1 or TMRE dyes
ATP production capacity
Mitochondrial morphology by electron microscopy or super-resolution imaging