The DVL3 Antibody is widely employed in molecular biology techniques to study DVL3’s role in cellular signaling, development, and disease mechanisms. Key applications include:
Western Blotting (WB): Detects DVL3 protein expression in lysates, with reported sensitivity in human, mouse, and rat samples .
Immunoprecipitation (IP): Used to isolate DVL3 for downstream analysis, such as studying protein-protein interactions .
Immunofluorescence (IF): Visualizes DVL3 localization in cells, including nuclear and cytoplasmic distribution .
Immunohistochemistry (IHC): Analyzes tissue-specific DVL3 expression, particularly in testicular and epithelial tissues .
| Antibody Source | Reactivity |
|---|---|
| Proteintech | Human, Mouse |
| DSHB (PCRP-DVL3) | Human |
| Cusabio | Human, Mouse, Rat |
| Cell Signaling | Human, Mouse, Rat |
DVL3’s role in cellular processes has been extensively studied:
Wnt Signaling: DVL3 shuttles between the cytoplasm and nucleus, regulating β-catenin stabilization and transcriptional activity .
Myogenesis: Knockdown of DVL3 impairs human myoblast proliferation and myogenic differentiation .
Spermatogenesis: DVL3 regulates the blood-testis barrier (BTB) and spermatid polarity via actin and microtubule cytoskeletons .
Cancer: Overexpression of DVL3 correlates with oncogenic signaling in certain tumors .
Figure 1: DVL3 Localization in Sertoli Cells
DVL3 localizes to the Sertoli cell–cell interface and basal ES/BTB in rat testes, as shown by immunofluorescence (green) .
DVL3 (Dishevelled-3) is a multivalent scaffold protein that plays an essential role in Wnt signaling during development . As one of three Dishevelled family members in mammals, DVL3 is involved in both canonical and non-canonical Wnt pathways, making it a crucial target for studying developmental processes, cancer progression, and various cellular signaling mechanisms. Research has revealed that DVL3 forms supermolecular complexes ranging from homodimers to well-defined structures of 0.4-2.0 MDa, with complex formation being stimulated by Wnt3a signaling within 30 minutes of exposure . This dynamic complex assembly is vital for proper signal transduction in the Wnt pathway.
DVL3 antibodies are extensively utilized in multiple research applications:
These applications allow researchers to study DVL3 protein expression levels, subcellular localization, interaction partners, and post-translational modifications in various experimental contexts .
When selecting a DVL3 antibody for your experiment, several critical factors should be considered:
Species reactivity: Confirm the antibody recognizes DVL3 from your experimental species. Available antibodies show reactivity with human, mouse, rat, hamster, and other species .
Specificity: Verify whether the antibody is specific to DVL3 or cross-reacts with other Dishevelled family members. Some antibodies, like #3218, specifically detect DVL3 without cross-reacting with DVL2 .
Application compatibility: Ensure the antibody is validated for your intended application. Check published literature and manufacturer data showing successful use in your application of interest .
Clonality: Choose between polyclonal antibodies (offering broader epitope recognition) or monoclonal antibodies (providing higher specificity) .
Target region: Consider whether your experiment requires detection of specific DVL3 domains or phosphorylation sites, especially for studies focused on Wnt signaling dynamics .
Review validation data, including western blot images and immunohistochemistry results, to confirm the antibody performs reliably in conditions similar to your experimental setup.
To study DVL3 supermolecular complex formation effectively, implement the following methodological approach:
Cell stimulation protocol: For studying Wnt-dependent complexes, stimulate cells with purified Wnt3a for specific time intervals (30 min, 1 hour) as this has been shown to induce progressive complex formation with detectable changes occurring within 30 minutes .
Sample preparation: Use steric-exclusion chromatography to separate DVL3-based complexes by size. This technique has successfully resolved complexes ranging from homodimeric DVL3 to supermolecular complexes of 0.4-2.0 MDa .
Immunoprecipitation optimization: For FLAG-tagged DVL3, use anti-FLAG immunoprecipitation followed by SDS-PAGE separation. For endogenous DVL3, use specialized antibodies at concentrations of 0.5-4.0 μg per 1.0-3.0 mg of total protein lysate .
Detection sensitivity: Employ fluorescence correlation microscopy to characterize the dynamic nature of DVL3 complexes in living cells, complementing biochemical approaches .
Controls: Include DVL1/2/3 knockdown controls to distinguish specific effects, as studies have shown that knockdown of each Dvl isoform affects complex formation differently. Notably, Dvl3 knockdown precludes complex formation, while Dvl1 knockdown specifically blocks Wnt3a-induced increases in complex size .
This multifaceted approach enables comprehensive analysis of DVL3's role in Wnt signaling complex assembly and dynamics.
DVL3 phosphorylation represents a critical regulatory mechanism within Wnt signaling cascades. To effectively analyze DVL3 phosphorylation:
Experimental design: Express FLAG-DVL3 in HEK293 cells with or without specific kinases of interest, followed by immunoprecipitation using anti-FLAG antibodies .
Phosphorylation analysis techniques:
Kinase modulation: Employ specific kinase inhibitors such as PF-670462 (CK1ε inhibitor at 10 μM) to evaluate the contribution of specific kinases to DVL3 phosphorylation patterns .
Functional correlation: Combine phosphorylation analysis with functional assays such as the TopFlash reporter assay in DVL1/2/3-null HEK293 T-REx cells transfected with DVL3 variants to connect phosphorylation status with Wnt signaling activation .
FlAsH labeling approach: For live-cell imaging of DVL3 conformational changes associated with phosphorylation, utilize the DVL3 FlAsH III sensor methodology with 500 nM FlAsH and 12.5 μM 1,2-ethanedithiol (EDT) as described in previous protocols .
This comprehensive approach links DVL3's phosphorylation state to its functional role in Wnt signaling cascades.
For precise subcellular localization analysis of DVL3:
Sample preparation protocol:
Antibody dilution optimization: For immunofluorescence/ICC applications, use DVL3 antibody at 1:50-1:500 dilution, with titration recommended for each experimental system .
Co-staining strategy: Implement co-staining with markers for specific subcellular compartments (nuclear, cytoplasmic, membrane, endosomal) to precisely define DVL3 localization under different signaling conditions.
Quantitative analysis approach: Apply semi-quantitative evaluation methods similar to those used in glioblastoma studies, where both signal intensity and subcellular distribution patterns are systematically quantified .
Physiological interpretation: Consider that DVL3 has been observed predominantly in the cytoplasm (97% of glioblastoma samples), with 44% of samples showing co-expression in the nucleus. Strong expression levels correlate significantly with nuclear localization (P = 6.33 × 10^-5) .
This methodology enables detection of key subcellular translocation events associated with DVL3's role in Wnt signaling activation.
To reduce non-specific binding in DVL3 antibody applications:
Western blotting optimization:
Immunoprecipitation refinement:
Immunohistochemistry considerations:
Fluorescence labeling specificity:
These methodological refinements significantly enhance signal-to-noise ratio and experimental reliability.
Several factors critically influence DVL3 antibody performance:
Buffer composition effects:
Sample preparation considerations:
Epitope accessibility variables:
Detection system compatibility:
Cell/tissue type variability:
Understanding these variables enables strategic optimization of experimental protocols for specific research questions.
A rigorous validation approach for DVL3 antibodies includes:
Genetic knockout/knockdown controls:
Signal specificity assessment:
Confirm expected molecular weight (78 kDa calculated, typically observed at 88-93 kDa due to post-translational modifications)
Verify absence of bands in DVL3-depleted samples
Evaluate cross-reactivity with other Dishevelled family members, particularly important since some antibodies specifically do not cross-react with DVL2
Functional validation approaches:
Recovery experiments:
Reintroduce tagged or untagged DVL3 into knockout systems and confirm signal restoration
Use multiple antibodies targeting different DVL3 epitopes to verify consistent detection patterns
Publication record evaluation:
This comprehensive validation approach ensures experimental results can be interpreted with high confidence.
For studying real-time Wnt signaling dynamics using DVL3 antibodies:
FlAsH-based sensor implementation:
Transfect cells with the DVL3 FlAsH III sensor construct
Label according to established protocols: incubate at 37°C for 1 hour with Hank's Balanced Salt Solution (HBSS) containing 500 nM FlAsH and 12.5 μM 1,2-ethanedithiol (EDT)
Reduce non-specific labeling with a 10-minute wash in HBSS containing 250 μM EDT
Complete with two HBSS washes before returning to DMEM medium
Live-cell imaging protocol:
Quantitative analysis approach:
Track DVL3 puncta formation and dynamics
Measure fluorescence correlation parameters to determine complex size changes
Analyze subcellular redistribution in response to pathway activation
FRAP/FRET techniques integration:
Combine with FRAP (Fluorescence Recovery After Photobleaching) to assess DVL3 mobility changes
Implement FRET systems to detect interactions between DVL3 and other Wnt pathway components
This approach provides dynamic insights into DVL3's role in Wnt signaling that complement static biochemical analyses.
DVL3 expression patterns in cancer contexts reveal important pathological insights:
Expression level analysis methodology:
Subcellular localization patterns:
Correlation with molecular profiles:
Tissue microarray applications:
Prognostic significance assessment:
Correlate expression patterns with patient outcome data
Compare with established molecular markers for the specific cancer type
This multifaceted approach provides insights into DVL3's potential role in cancer pathogenesis and progression.
Post-translational modifications (PTMs) of DVL3 significantly impact antibody recognition:
Phosphorylation effects:
Extensive phosphorylation alters DVL3's apparent molecular weight from the calculated 78 kDa to observed 88-93 kDa in western blots
Kinase activity (particularly CK1ε) induces mobility shifts that may affect epitope accessibility
Use of phosphatase treatment as a control can help distinguish phosphorylation-dependent epitope masking
Ubiquitination considerations:
Ubiquitination state affects DVL3 stability and complex formation
High molecular weight smears in western blots may indicate ubiquitinated forms
Deubiquitinating enzyme inhibitors may be necessary to preserve these modifications during sample preparation
Protein conformation influence:
Antibody selection strategy:
For PTM-specific studies, select antibodies with epitopes distant from known modification sites
Consider using multiple antibodies targeting different regions to obtain comprehensive detection
Experimental design adaptation:
Understanding these PTM effects is crucial for accurate interpretation of experimental results, particularly in signaling studies where DVL3's modification state directly reflects pathway activity.
For investigating signaling cross-talk involving DVL3:
Co-immunoprecipitation strategy:
Stimulation experimental design:
Compare DVL3 complex composition under single vs. multiple pathway stimulation
Track temporal dynamics of complex assembly/disassembly following sequential pathway activation
Analyze how inhibition of one pathway affects DVL3's role in another
Proximity ligation assay application:
Visualize endogenous protein-protein interactions in situ
Quantify interaction frequency in different subcellular compartments
Compare interaction patterns across normal vs. pathological conditions
Combinatorial knockdown approach:
Post-translational modification cross-regulation:
Investigate how kinases from different pathways affect DVL3 phosphorylation patterns
Analyze whether these modifications alter DVL3's scaffold function across pathways
This integrated approach reveals DVL3's role as a potential signaling hub coordinating multiple cellular processes.
When investigating DVL3 in primary patient samples:
Tissue preservation protocol optimization:
For immunohistochemistry applications, optimize fixation timing to prevent epitope masking
Consider specialized preservation methods for phosphorylated DVL3 detection
Antigen retrieval method selection:
Antibody dilution adaptation:
Comparative analysis framework:
RNA-protein correlation assessment:
Perform parallel analysis of DVL3 mRNA expression (RT-qPCR, RNA-seq)
Compare protein localization with transcript abundance
Investigate potential post-transcriptional regulatory mechanisms
This methodological approach enables reliable analysis of DVL3's contribution to human pathologies while accounting for sample heterogeneity.
For comparative studies of DVL family members:
Specificity verification approach:
Structural homology considerations:
DVL family members share conserved domains (DIX, PDZ, DEP) but differ in specific sequences
Target unique regions for differential detection
Account for isoform-specific variations that may affect antibody binding
Expression level normalization:
Functional redundancy analysis:
Implement knockdown/knockout of individual DVL members to assess specific functions
Compare subcellular distributions of different DVL proteins under identical conditions
Analyze differential complex formation (DVL1 and DVL3 overexpression stimulates formation of large supermolecular complexes, while DVL2 shows different patterns)
Systemic approach implementation:
Design experiments to simultaneously detect all DVL proteins in the same samples
Compare post-translational modification patterns across family members
Correlate with functional readouts to distinguish specific roles
This systematic comparison reveals both overlapping and distinct functions of DVL family members in Wnt signaling and beyond.