DTL (denticleless E3 ubiquitin protein ligase homolog) is a substrate-specific adapter of the DCX (DDB1-CUL4-X-box) E3 ubiquitin-protein ligase complex that plays critical roles in cell cycle control, DNA damage response, and translesion DNA synthesis. In humans, the canonical form consists of 730 amino acid residues with a molecular mass of approximately 79.5 kDa . DTL is also known by several synonyms including DCAF2, L2DTL, RAMP, denticleless protein homolog, DDB1- and CUL4-associated factor 2, and CDT2 . The protein contains characteristic WD40 repeat domains and localizes to the membrane, nucleus, and cytoplasm . DTL has gained significant research interest due to its involvement in multiple cancers and potential value as a diagnostic and prognostic biomarker .
DTL antibodies are utilized for multiple research applications, with the most common being:
Western Blotting (WB): For detecting DTL protein expression levels in cell and tissue lysates
Immunohistochemistry (IHC): For visualizing DTL expression patterns in tissue sections
Immunoprecipitation (IP): For isolating DTL protein complexes and studying protein-protein interactions
Immunofluorescence (IF): For subcellular localization studies
Flow Cytometry (FCM): For quantitative analysis of DTL expression in cell populations
The selection of appropriate antibody depends on the specific application, with different suppliers offering antibodies validated for different techniques. For complex experiments involving multiple detection methods, researchers should select antibodies validated across all required applications.
When validating DTL antibodies, researchers should follow these methodological steps:
Positive and negative controls: Use cell lines with known DTL expression levels (high in placenta and testis tissues) versus low-expression tissues (skeletal muscle) .
Knockdown/knockout validation: Implement DTL knockdown using validated shRNA sequences (e.g., GCCTAGTAACAGTAACGAGTA, CTGGTGAACTTAAACTTGTTA, or GCTCCCAATATGGAACATGTA) to confirm antibody specificity.
Western blot analysis: Verify single band detection at the expected molecular weight (79.5 kDa) .
Cross-reactivity assessment: If working with non-human models, confirm reactivity with the species of interest, as DTL orthologs exist in mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken species .
Application-specific validation: For IHC applications, include known positive tissues and optimize staining parameters using the staining index (SI) calculation: SI = positive staining score (0-4 points) × staining intensity score (0-3 points) .
To investigate DTL's role in cancer progression, researchers should implement a multi-faceted approach:
Comparative expression analysis: Use DTL antibodies for IHC or IF to quantify expression levels across normal tissues, primary tumors, and metastatic samples. Implement the scoring system where the staining index (SI) is calculated by multiplying the positive staining score (percentage of positive cells on a 0-4 scale) by the staining intensity score (0-3 scale) .
Correlation with clinical parameters: Analyze DTL expression in relation to clinical features such as tumor stage, grade, and patient outcomes. Divide samples into high and low DTL expression groups based on median SI scores (e.g., median of 10 points) for statistical analyses .
Co-localization studies: Use dual immunofluorescence to assess DTL co-localization with other cancer-related proteins. For example, examining the relationship between DTL expression and immune cell infiltration markers like CD3 can provide insights into the tumor microenvironment .
Functional validation: Implement DTL overexpression or knockdown in cell lines, followed by cell proliferation assays (e.g., BrdU incorporation), cell cycle analysis, and genomic stability assessments to determine the functional consequences of altered DTL expression .
Downstream signaling analysis: Use DTL antibodies in combination with antibodies against known interacting partners (e.g., PDCD4, RUVBL1) to elucidate the molecular mechanisms through which DTL promotes cancer progression .
To effectively study DTL-mediated protein ubiquitination, researchers should follow these methodological approaches:
Co-immunoprecipitation (Co-IP) assays: Use DTL antibodies to pull down protein complexes, followed by immunoblotting for potential target proteins. This approach has successfully identified interactions between DTL and proteins like PDCD4 .
Affinity-purification mass spectrometry: This comprehensive approach can identify novel DTL interaction partners. Transfect cells with Flag-DTL plasmid, immunoprecipitate using Flag magnetic affinity resin, separate by SDS-PAGE, and analyze excised bands by mass spectrometry .
Ubiquitination assays: To detect ubiquitination of target proteins:
Co-transfect cells with plasmids expressing DTL and the target protein
Treat cells with proteasome inhibitors (e.g., MG132) to prevent degradation of ubiquitinated proteins
Immunoprecipitate the target protein and immunoblot with anti-ubiquitin antibodies
Alternatively, use ubiquitin mutants (K48R or K63R) to distinguish between different ubiquitination types
Domain mapping experiments: Generate truncated fragments of DTL (e.g., constructs containing only the WD40 domains) to identify regions responsible for target protein interactions. This approach revealed that the WD40 domains (amino acids 35-398) are essential for DTL's interaction with PDCD4 .
In vitro ubiquitination assays: Reconstitute the ubiquitination reaction using purified components (E1, E2, DTL-containing E3 complex, ubiquitin, and substrate) to directly demonstrate DTL-mediated ubiquitination.
To investigate DTL's role in DNA damage response (DDR) pathways, researchers should employ these methodological approaches:
Radiation-induced damage models: Expose cells to radiation treatment and analyze changes in DTL expression and localization using DTL antibodies. This approach revealed that RUVBL1 ubiquitination by DTL promotes the formation of RUVBL1/2-β-catenin transcription complexes following radiation treatment .
Co-localization with DDR markers: Perform immunofluorescence to assess co-localization of DTL with DNA damage markers (γH2AX) and repair proteins at different time points after damage induction.
Chromatin immunoprecipitation (ChIP): Use DTL antibodies for ChIP assays to determine if DTL associates with chromatin at damaged sites.
Protein complex analysis: Implement immunoprecipitation with DTL antibodies followed by mass spectrometry to identify DDR-related interaction partners under normal and damage-induced conditions.
Functional assays: Assess the impact of DTL knockdown or overexpression on:
Cell cycle checkpoint analysis: Use flow cytometry with DTL antibodies to analyze the role of DTL in radiation-induced G2/M checkpoint activation, as DTL has been identified as an essential component of this checkpoint .
For optimal Western blotting with DTL antibodies, researchers should follow these methodological recommendations:
Sample preparation:
Gel electrophoresis:
Transfer and blocking:
Transfer to PVDF membrane at 100V for 90 minutes in cold transfer buffer
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Antibody incubation:
Primary antibody dilution: 1:1000 to 1:2000 (optimize based on specific antibody)
Incubate overnight at 4°C
Wash 3x with TBST
Secondary antibody: Use HRP-conjugated antibody at 1:5000 dilution for 1 hour at room temperature
Detection:
Troubleshooting:
If detecting multiple bands, increase washing stringency and optimize antibody dilution
If signal is weak, increase protein loading or antibody concentration
For low abundance in certain tissues, consider immunoprecipitation before Western blotting
When manipulating DTL expression levels, include these controls to ensure experimental validity:
Knockdown controls:
Non-targeting shRNA/siRNA control
Multiple DTL-targeting shRNA sequences to control for off-target effects (recommended sequences: GCCTAGTAACAGTAACGAGTA, CTGGTGAACTTAAACTTGTTA, GCTCCCAATATGGAACATGTA)
Rescue experiments with shRNA-resistant DTL constructs
qRT-PCR and Western blot validation of knockdown efficiency
Overexpression controls:
Functional controls:
Pathway validation:
For accurate detection and quantification of DTL in tissue samples, researchers should:
Immunohistochemistry (IHC) protocol:
Use paraffin-embedded or frozen tissue sections (5-7 μm thickness)
Perform antigen retrieval in citrate buffer (pH 6.0)
Block endogenous peroxidase with 3% H₂O₂
Block with 5% BSA or serum
Incubate with anti-DTL primary antibody overnight at 4°C
Implement a standardized scoring system where:
Positive staining score: 0 (0-5%), 1 (5-25%), 2 (26-50%), 3 (51-75%), 4 (>75%)
Staining intensity score: 0 (no staining), 1 (weak/light yellow), 2 (moderate/bright yellow), 3 (strong/brown)
Staining index (SI) = positive staining score × intensity score
Categorize as high or low DTL expression based on median SI value (e.g., 10 points)
Immunofluorescence (IF) for co-localization studies:
After staining with primary antibodies (e.g., anti-DTL and anti-CD3)
Incubate with appropriate secondary antibodies (anti-rabbit-Cy3 for DTL, anti-mouse-FITC for CD3)
Counterstain nuclei with DAPI
Capture images using fluorescence microscopy
Quantify percentage of positive cells and analyze relationship between DTL expression and other markers
Tissue microarray (TMA) analysis:
For high-throughput analysis of multiple samples
Ensure inclusion of normal tissue controls
Apply the standardized scoring system described above
Use digital pathology software for automated quantification when possible
To properly analyze associations between DTL expression and clinical outcomes, researchers should follow these methodological approaches:
To investigate DTL's role in tumor immunology, researchers should employ these methodological approaches:
Immune infiltration analysis:
Perform multiplex immunofluorescence staining for DTL and immune cell markers (e.g., CD3 for T cells)
Quantify immune cell populations in high vs. low DTL-expressing regions
Analyze the relationship between DTL expression and the percentage of CD3-positive cells by dividing samples into high and low CD3 groups according to median values (e.g., 10%)
Immunotherapy response prediction:
Analyze DTL expression in pre-treatment biopsies from immunotherapy responders vs. non-responders
Utilize datasets containing DTL transcriptomic and genomic profiling with immunotherapy outcomes (e.g., GEO: GSE78220, GEO: GSE67501, IMvigor210)
Compare DTL expression between responders and non-responders to checkpoint blockade (anti-PDL1, anti-PD1)
In vitro immune interaction models:
Co-culture tumor cells with varying DTL expression levels with immune cells
Assess changes in immune activation markers and cytokine production
Evaluate the effect of DTL knockdown or overexpression on immune cell recruitment and function
Pathway analysis:
Investigate correlations between DTL expression and immune-related signaling pathways
Use GSEA to identify immune pathways associated with DTL expression levels
Analyze the relationship between DTL-mediated ubiquitination and immune response modulation
When facing inconsistent results in DTL research, implement these methodological approaches:
Antibody validation and standardization:
Verify antibody specificity through knockdown/knockout controls
Compare multiple commercial antibodies for consistency
Use the same antibody lot number throughout a study
Include positive control samples with known DTL expression
Experimental design considerations:
Control for cell confluence and passage number in cell line studies
Standardize tissue collection, processing, and storage protocols
Implement blinded scoring in IHC/IF studies
Use technical and biological replicates
Context-specific expression analysis:
Account for tumor heterogeneity by analyzing multiple regions
Consider microenvironmental factors that might influence DTL expression
Analyze DTL expression in relation to cell cycle phase
Evaluate post-translational modifications that might affect antibody recognition
Statistical approaches:
Perform power calculations to ensure adequate sample size
Use appropriate statistical tests based on data distribution
Implement multiple testing corrections
Report effect sizes alongside p-values
Consider Bayesian approaches for integrating conflicting data
Researchers should consider these emerging technologies to enhance DTL research:
Single-cell analysis:
Single-cell proteomics to analyze DTL expression heterogeneity within tumors
Single-cell RNA-seq combined with protein expression (CITE-seq) to correlate DTL transcription and translation
Spatial transcriptomics to map DTL expression within the tumor microenvironment
Advanced imaging techniques:
Super-resolution microscopy for detailed subcellular localization
Live-cell imaging with fluorescently tagged DTL to track dynamics during cell cycle and DNA damage
Multiplexed ion beam imaging (MIBI) or cyclic immunofluorescence for simultaneous detection of multiple markers
Protein interaction mapping:
Proximity labeling techniques (BioID, APEX) to identify the DTL interactome in living cells
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map protein interaction domains
Cryo-electron microscopy to determine 3D structures of DTL-containing complexes
Functional genomics:
CRISPR-Cas9 screens to identify synthetic lethal interactions with DTL
CRISPR base editing to introduce specific mutations in DTL domains
Degron tagging for acute depletion of DTL protein
Researchers face several challenges in developing specific DTL antibodies:
Structural complexity:
Post-translational modifications:
Cross-reactivity concerns:
Technical limitations:
DTL antibody research is revealing several promising therapeutic directions:
Cancer diagnostics and prognostics:
Targeted therapy approaches:
Immunotherapy enhancement:
Radiotherapy sensitization:
Through continued research using high-quality DTL antibodies, these therapeutic applications may translate into clinically meaningful advances for cancer patients.