DVL1P1 is a paralog of the Dishevelled (DVL) family, which mediates Wnt signaling—a pathway critical for embryonic development, cell polarity, and proliferation . While DVL1 (a closely related isoform) directly interacts with Frizzled receptors to regulate β-catenin stabilization , DVL1P1’s exact mechanistic role remains less characterized. Studies suggest it may act as a signaling intermediary, though its functional overlap with other DVL isoforms requires further investigation .
Although no direct studies using this specific antibody are cited in the provided sources, research on related DVL proteins highlights their broader biological relevance. For example:
Immune Regulation: DVL1 knockout mice exhibit altered gut microbiota and increased lymphoid aggregates in the colon, suggesting a role in immune-epithelial crosstalk .
Pathological Implications: Dysregulated DVL signaling is linked to cancers and neurodegenerative diseases, underscoring the importance of reagents like this antibody for mechanistic studies .
The FITC-conjugated DVL1P1 antibody (A20006, AFG Scientific) is priced at $225 (50 µl) and $330 (100 µl) . Comparable antibodies targeting DVL1 (not DVL1P1) include:
| Product (Conjugate) | Host | Clonality | Price (200 µg/ml) |
|---|---|---|---|
| DVL1P1-FITC | Rabbit | Polyclonal | $225–$330 |
| DVL1-FITC (sc-8025) | Mouse | Monoclonal | $330 |
Specificity: While the antibody is validated for ELISA, its performance in advanced applications (e.g., immunofluorescence or Western blot) remains unconfirmed in the provided data .
Research Gaps: No peer-reviewed studies directly utilizing this conjugate were identified. Further work could explore its utility in:
Mapping DVL1P1 expression patterns in Wnt-driven cancers.
Investigating cross-reactivity with DVL isoforms in model organisms.
DVL1P1 (Dishevelled Segment Polarity Protein 1 Pseudogene 1) is a protein that may play a significant role in the signal transduction pathway mediated by multiple Wnt genes . It is structurally related to the DVL1 protein, which participates in Wnt signaling by binding to the cytoplasmic C-terminus of frizzled family members and transducing the Wnt signal to downstream effectors . DVL1 plays roles in both canonical and non-canonical Wnt signaling pathways, which are critical for embryonic development, tissue homeostasis, and can be dysregulated in various diseases including cancer.
While DVL1 has been extensively characterized, research on DVL1P1 continues to evolve. The protein contains sequence from amino acids 152-173 that serves as an immunogen for antibody production . Understanding DVL1P1's function provides important insights into Wnt signaling regulation and potential therapeutic targets.
The DVL1P1 Antibody, FITC conjugated, is typically a polyclonal antibody raised in rabbits against a peptide sequence from the human putative segment polarity protein dishevelled homolog DVL1P1 (amino acids 152-173) . The antibody is supplied with the following specifications:
| Parameter | Specification |
|---|---|
| Host Species | Rabbit |
| Clonality | Polyclonal |
| Target | Human DVL1P1 |
| Immunogen | Peptide sequence from Human DVL1P1 protein (152-173AA) |
| Conjugate | FITC (Fluorescein isothiocyanate) |
| Applications | ELISA, potentially Western Blot |
| Isotype | IgG |
| Form | Liquid |
| Buffer | 50% Glycerol, 0.01M PBS, pH 7.4, 0.03% Proclin 300 |
| Storage | -20°C or -80°C; avoid repeated freeze-thaw cycles |
| Purification Method | Antigen Affinity Purified |
| UniProt ID | P54792 |
These technical specifications are essential for researchers to evaluate the suitability of the antibody for their specific experimental requirements .
Proper storage and handling of DVL1P1 Antibody, FITC conjugated is crucial for maintaining its activity and specificity:
Storage temperature: Upon receipt, store at -20°C or -80°C . The lower temperature of -80°C is recommended for long-term storage.
Aliquoting: To minimize freeze-thaw cycles, divide the antibody into small aliquots before freezing, especially if you won't use the entire volume at once.
Freeze-thaw cycles: Avoid repeated freeze-thaw cycles as this can lead to denaturation and loss of activity .
Light sensitivity: As FITC is a fluorophore sensitive to light, the antibody should be protected from prolonged exposure to light during handling and storage to prevent photobleaching.
Buffer conditions: The antibody is supplied in a buffer containing 50% glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as a preservative . This formulation helps maintain stability during freezing.
Working dilutions: Prepare working dilutions immediately before use and discard any unused diluted antibody.
Contamination prevention: Use sterile techniques when handling to prevent microbial contamination.
Proper storage and handling significantly impact experimental outcomes and reproducibility. Researchers should carefully monitor storage conditions to ensure antibody performance remains consistent across experiments.
Optimizing protocols for fluorescence-based applications with DVL1P1 Antibody, FITC conjugated requires careful consideration of several parameters:
For Immunofluorescence Microscopy:
Fixation method optimization: Compare paraformaldehyde (4%) versus methanol fixation to determine which better preserves epitope recognition while maintaining cellular architecture.
Permeabilization conditions: Test Triton X-100 (0.1-0.5%), saponin (0.1%), or digitonin (10-50 μg/ml) to identify optimal conditions that allow antibody access while preserving subcellular structures.
Blocking parameters: Use 5-10% normal serum (from the species of the secondary antibody if used in a detection system) with 1-3% BSA to minimize background signal.
Antibody dilution optimization: Test serial dilutions (typically starting at 1:50-1:200) to determine the optimal signal-to-noise ratio .
Counterstaining: DAPI (1 μg/ml) for nuclear visualization can provide context for DVL1P1 localization, which may be important for Wnt signaling studies.
For Flow Cytometry:
Cell preparation: Single-cell suspensions with viability >90% are essential for reliable results.
Fixation and permeabilization: For intracellular targets, use commercial fixation/permeabilization kits compatible with FITC fluorescence.
Antibody titration: Test dilutions from 1:10 to 1:500 to determine optimal staining conditions .
Controls: Include unstained cells, isotype controls (FITC-conjugated rabbit IgG), and positive controls (cell lines known to express DVL1P1).
Compensation: If performing multicolor flow cytometry, proper compensation is essential due to FITC spectral overlap with other fluorophores.
The distinct advantage of FITC conjugation is that it eliminates the need for secondary antibody incubation, reducing potential cross-reactivity issues and simplifying protocols.
Cross-reactivity between DVL1P1 and related proteins presents a significant challenge for specific detection. Researchers can address this through several methodological approaches:
This systematic approach ensures confidence in experimental results when studying specific family members in the dishevelled protein family.
Designing effective multiplex immunofluorescence experiments with DVL1P1 Antibody, FITC conjugated requires careful planning to maximize information while avoiding technical artifacts:
Spectral compatibility: FITC excites at ~495 nm and emits at ~519 nm. When selecting additional fluorophores, choose those with minimal spectral overlap such as:
Texas Red (excitation: ~589 nm, emission: ~615 nm)
Cy5 (excitation: ~650 nm, emission: ~670 nm)
Pacific Blue (excitation: ~410 nm, emission: ~455 nm)
Antibody compatibility:
Host species considerations: When using multiple primary antibodies, select those raised in different host species to enable specific secondary detection
For directly conjugated antibodies like DVL1P1-FITC, ensure other conjugated antibodies don't cross-react with your target proteins
Staining sequence optimization:
Sequential staining protocols may be necessary if antibodies are from the same host species
Test order of antibody application to determine if one antibody blocks access to other epitopes
Signal amplification strategies: For low-abundance targets that might be co-detected with DVL1P1, consider:
Tyramide signal amplification (TSA) for other markers
Use of quantum dots for brighter, more stable fluorescence
Note that DVL1P1-FITC signal cannot be amplified through secondary antibodies due to direct conjugation
Image acquisition parameters:
Use sequential scanning to minimize bleed-through
Standardize exposure times and gain settings across experimental conditions
Consider spectral unmixing algorithms for closely overlapping fluorophores
Controls for multiplex experiments:
Single-stained controls for each fluorophore
Fluorescence minus one (FMO) controls
Isotype controls for each conjugated antibody
DVL1P1-specific considerations:
This comprehensive approach enables robust multiplex experiments investigating DVL1P1 in complex cellular contexts.
The DVL1P1 Antibody, FITC conjugated provides unique opportunities for investigating Wnt signaling dynamics through various methodological approaches:
Live-cell imaging of Wnt pathway activation:
Transfect cells with fluorescently-tagged Wnt pathway components (e.g., RFP-tagged β-catenin)
Permeabilize cells with a gentle detergent to allow antibody entry while maintaining viability
Use DVL1P1-FITC to visualize DVL1P1 localization during Wnt stimulation
Capture images at defined intervals following Wnt ligand addition (e.g., 0, 15, 30, 60 minutes)
Quantify co-localization coefficients between DVL1P1 and other pathway components
Quantitative analysis of DVL1P1 in signalosome formation:
Wnt ligand binding triggers formation of signalosomes at the plasma membrane
Use super-resolution microscopy (STORM, PALM) with DVL1P1-FITC to quantify:
Signalosome size
Protein stoichiometry within signalosomes
Temporal dynamics of assembly/disassembly
Differential response to canonical vs. non-canonical Wnt ligands:
Compare DVL1P1 localization following treatment with:
Wnt3a (canonical pathway activator)
Wnt5a (non-canonical pathway activator)
Quantify differences in cytoplasmic vs. membrane-associated DVL1P1-FITC signal
Phosphorylation state analysis:
DVL proteins undergo phosphorylation upon Wnt activation
Combine DVL1P1-FITC with phospho-specific antibodies (using a compatible fluorophore)
Create a temporal map of phosphorylation events during signaling
Interaction with downstream effectors:
Perform proximity ligation assays (PLA) using DVL1P1-FITC and antibodies against potential interacting partners
Quantify interaction events during pathway activation/inhibition
Pharmacological manipulation:
Apply pathway modulators including:
GSK3β inhibitors (e.g., CHIR99021)
Tankyrase inhibitors (e.g., XAV939)
Porcupine inhibitors (e.g., LGK974)
Monitor DVL1P1 localization and abundance changes using the FITC-conjugated antibody
Correlate these changes with downstream signaling outcomes
This multi-faceted approach leverages the direct FITC conjugation to minimize experimental perturbation while maximizing dynamic information acquisition.
Comprehensive validation of DVL1P1 Antibody, FITC conjugated is essential before implementing it in critical research applications. A systematic validation approach should include:
Specificity validation:
Western blot analysis: Confirm antibody detects a single band of appropriate molecular weight in cell/tissue lysates expressing DVL1P1
Immunoprecipitation followed by mass spectrometry: Verify antibody pulls down DVL1P1 without significant off-target proteins
Genetic ablation controls: Test antibody in CRISPR/Cas9 knockout or siRNA knockdown systems
Recombinant protein detection: Test against purified DVL1P1 and related family members
Sensitivity assessment:
Titration experiments: Determine limit of detection using serial dilutions of recombinant protein
Signal-to-noise ratio quantification: Calculate across multiple applications and sample types
Comparison with alternative detection methods: qPCR for mRNA expression correlation
Performance across applications:
Application-specific validation: For each intended application (ELISA, flow cytometry, immunofluorescence), perform dedicated validation experiments
Sample preparation optimization: Test multiple fixation and permeabilization protocols to maximize signal while preserving epitope accessibility
Antibody dilution series: Determine optimal working concentration for each application
FITC conjugation quality control:
Degree of labeling (DOL) determination: Calculate fluorophore-to-protein ratio using spectrophotometric methods
Free dye assessment: Confirm absence of unconjugated FITC which could increase background
Functional impact assessment: Compare binding efficacy of conjugated vs. unconjugated antibody
Reproducibility testing:
Lot-to-lot consistency: Test multiple antibody lots on standardized samples
Inter-laboratory validation: If possible, exchange protocols and samples with collaborating labs
Long-term stability assessment: Test antibody performance after storage under recommended conditions for varying time periods
Context-specific validations:
Cell type specificity: Verify performance across relevant cell lines and primary cells
Tissue-specific artifacts: Assess background and specificity in tissues of interest
Species cross-reactivity: Although labeled for human reactivity, test cross-reactivity with model organism samples if relevant to research
Thorough documentation of these validation steps provides confidence in experimental results and facilitates troubleshooting when unexpected results occur.
The DVL1P1 Antibody, FITC conjugated offers valuable applications in cancer research, particularly given the importance of Wnt signaling in oncogenesis and therapy resistance:
Diagnostic and prognostic biomarker exploration:
Analyze DVL1P1 expression patterns across cancer subtypes using tissue microarrays
Correlate expression levels with clinical outcomes and treatment responses
Develop quantitative scoring systems based on fluorescence intensity and subcellular localization patterns
Integration with anti-FITC CAR-T cell therapy platforms:
The FITC conjugation creates opportunities for targeted immunotherapy approaches
Anti-FITC CAR T cells can recognize cancer cells bound with FITC-labeled antibodies
Research workflow would include:
a. Validating DVL1P1 surface exposure in cancer cells
b. Optimizing antibody concentration for CAR-T cell recognition
c. Assessing cytotoxicity in co-culture systems
d. Evaluating potential for on-target/off-tumor effects
Wnt pathway inhibitor screening:
Use high-content imaging with DVL1P1-FITC to screen compound libraries
Quantify changes in DVL1P1 localization, degradation, or signalosome formation
Correlate DVL1P1 dynamics with downstream pathway activation markers
Identify compounds that specifically modulate DVL1P1 without affecting related family members
Resistance mechanism investigations:
Compare DVL1P1 expression and localization in therapy-sensitive versus resistant cells
Use time-lapse imaging to track DVL1P1-FITC during development of resistance
Combine with other pathway markers to identify compensatory signaling networks
Cancer stem cell (CSC) characterization:
Wnt signaling is crucial for maintaining cancer stem cell populations
Use DVL1P1-FITC to identify and isolate cells with distinct Wnt signaling states
Combine with CSC markers (CD44, CD133, ALDH) in flow cytometry to correlate DVL1P1 levels with stemness
Translational research applications:
Develop image analysis algorithms for quantifying DVL1P1 in clinical samples
Standardize reporting methods for pathologists evaluating DVL1P1 expression
Create reference datasets for interpreting DVL1P1 alterations in patient samples
The direct FITC conjugation provides advantages for multiparameter analyses in complex tumor microenvironments and enables novel therapeutic approaches leveraging antibody targeting specificity.
When experiments with DVL1P1 Antibody, FITC conjugated yield suboptimal results, a systematic troubleshooting approach is essential:
No signal or weak signal issues:
Antibody integrity: Check for signs of denaturation (precipitation, unusual color)
Epitope accessibility: Test alternative fixation methods; some fixatives may mask the epitope
Target expression level: Verify DVL1P1 expression in your sample by RT-PCR or Western blot
Photobleaching: Minimize exposure to light during all steps; use anti-fade mounting media
Working concentration: Test higher antibody concentrations (1:50-1:100 range)
Detection system sensitivity: Use more sensitive cameras/detectors or increase exposure time
High background or non-specific signal:
Inadequate blocking: Increase blocking time/concentration or try alternative blocking reagents
Autofluorescence: Include unstained controls; use tissues/cells known to have low autofluorescence
Fixative-induced fluorescence: Try alternative fixation methods
Non-specific binding: Include additional wash steps and increase wash buffer stringency
Cross-reactivity: Pre-absorb antibody with related proteins (DVL1, DVL2, DVL3)
Over-concentration: Dilute antibody further if background is uniformly high
Inconsistent results across experiments:
Sample variability: Standardize sample collection, processing, and storage
Antibody storage issues: Aliquot antibody to avoid repeated freeze-thaw cycles
Protocol timing variations: Time each step precisely across experiments
Lot-to-lot variation: Request information about lot-specific validation from manufacturer
Environmental factors: Control room temperature, humidity, and light exposure
Application-specific troubleshooting:
Flow cytometry issues:
Dead cell exclusion: Include viability dye to gate out dead cells
Cell clumping: Improve single-cell suspension preparation
Compensation problems: Run single-color controls for accurate compensation
Immunofluorescence issues:
Focus problems: Ensure proper mounting and appropriate coverslip thickness
Fading signal: Use anti-fade mounting media and minimize exposure during imaging
Uneven staining: Check for air bubbles or incomplete reagent coverage
Experimental design refinements:
Positive control inclusion: Include samples known to express DVL1P1
Negative control optimization: Use isotype controls (FITC-conjugated rabbit IgG)
Alternative antibody comparison: Test an unconjugated DVL1P1 antibody with secondary detection
Technical validation tests:
Peptide competition assay: Pre-incubate antibody with immunizing peptide to confirm specificity
Fluorophore activity test: Verify FITC fluorescence using standard fluorescence measurement
Antibody binding verification: Perform ELISA against immunizing peptide
This methodical approach helps isolate the source of experimental problems and guides efficient resolution strategies.