TRABD2B Antibody, HRP conjugated is a polyclonal antibody that specifically recognizes the human TRABD2B protein, also known as TIKI2 or Metalloprotease TIKI2. The antibody is conjugated to Horseradish Peroxidase (HRP), an enzyme that catalyzes reactions producing a detectable signal in various immunoassay applications . This antibody serves as a valuable research tool for studying TRABD2B's biological functions, particularly its role in regulating the Wnt signaling pathway.
TRABD2B Antibody, HRP conjugated is produced by immunizing rabbits with a recombinant fragment of human TRABD2B protein, typically amino acids 195-405. The resulting polyclonal antibody is then affinity-purified and conjugated to HRP to enable direct detection without requiring secondary antibodies . This conjugation significantly streamlines experimental workflows while maintaining specificity for the target protein.
Understanding the target protein is essential for proper application of the TRABD2B Antibody, HRP conjugated in research contexts.
TRABD2B serves as a negative regulator of the Wnt signaling pathway by functioning as a metalloprotease. Its primary mechanism involves:
Mediating the cleavage of the 8 N-terminal residues of specific Wnt proteins
Following cleavage, Wnt proteins become oxidized and form large disulfide-bond oligomers
This oxidation and oligomerization leads to inactivation of the Wnt proteins
The protein shows substrate specificity, being able to cleave WNT3A and WNT5, but not WNT11
TRABD2B is notably required for head formation during development and is enriched in heart, kidney, and adipose tissues, as reflected in one of its alternative names (HKAT) .
The direct HRP conjugation of this antibody makes it particularly suitable for certain research applications where direct detection is advantageous.
TRABD2B Antibody, HRP conjugated is primarily recommended for ELISA applications . In this context, the antibody can bind specifically to TRABD2B protein immobilized on a solid support, and the conjugated HRP enzyme catalyzes a colorimetric reaction for detection and quantification. This application enables researchers to:
Detect the presence of TRABD2B in complex biological samples
Quantify TRABD2B expression levels
Study protein-protein interactions involving TRABD2B
While the primary verified application is ELISA, similar HRP-conjugated antibodies are often suitable for Western Blot applications as well. In Western Blotting, the antibody can detect TRABD2B protein that has been separated by gel electrophoresis and transferred to a membrane . The direct HRP conjugation eliminates the need for secondary antibody incubation, simplifying the protocol and potentially reducing background signal.
The TRABD2B Antibody, HRP conjugated serves as an important tool for investigating:
Wnt signaling pathway dysregulation in various diseases
Mechanisms of Wnt protein processing and regulation
TRABD2B expression patterns in different tissues and cell types
Potential therapeutic targets in the Wnt pathway
When working with TRABD2B Antibody, HRP conjugated, several experimental considerations should be taken into account to ensure optimal results.
When using this antibody, appropriate experimental controls should be included:
Positive controls: Samples known to express TRABD2B, such as specific cell lines (DU 145, HT-29, HeLa cells have been reported to express TRABD2B)
Negative controls: Samples known not to express TRABD2B or samples where the protein has been knocked down
Background controls: To assess non-specific binding of the antibody
For visualizing the HRP activity in various applications, several detection methods can be employed:
Colorimetric detection using substrates like TMB (3,3',5,5'-Tetramethylbenzidine)
Chemiluminescent detection using enhanced chemiluminescence (ECL) substrates
Chemifluorescent detection using appropriate substrates
The choice of detection method depends on the specific research requirements such as sensitivity needs, equipment availability, and experimental design.
Understanding how TRABD2B Antibody, HRP conjugated compares to related antibodies provides context for selecting the appropriate research reagent.
Several variants of TRABD2B antibodies are commercially available, each with different conjugations and applications:
| Antibody Variant | Conjugation | Primary Applications | Host |
|---|---|---|---|
| TRABD2B Antibody, HRP conjugated | HRP | ELISA | Rabbit |
| TRABD2B Antibody, FITC conjugated | FITC | Flow cytometry, immunofluorescence | Rabbit |
| TRABD2B Antibody, Biotin conjugated | Biotin | ELISA, immunohistochemistry | Rabbit |
| TRABD2B Antibody, unconjugated | None | Western blot, ELISA, immunofluorescence, immunohistochemistry | Rabbit |
This comparison highlights the specialized nature of the HRP-conjugated variant for applications where enzymatic detection is advantageous .
HRP conjugation is a common modification for many antibodies used in research. Some characteristics of HRP-conjugated antibodies include:
Direct detection without secondary antibodies
Reduced background in some applications
Shorter protocols due to elimination of secondary antibody steps
Potential for higher sensitivity in certain detection systems
The TRABD2B Antibody, HRP conjugated shares these general characteristics with other HRP-conjugated antibodies, such as HRP-conjugated anti-tag antibodies like anti-6X His tag or anti-HA tag antibodies .
The TRABD2B Antibody, HRP conjugated has significant relevance in various research contexts related to developmental biology, cancer research, and molecular signaling.
The Wnt signaling pathway plays crucial roles in:
Embryonic development
Tissue homeostasis
Cancer progression
Stem cell maintenance
As TRABD2B functions as a negative regulator of this pathway, the TRABD2B Antibody, HRP conjugated provides a valuable tool for studying these processes .
Research utilizing TRABD2B Antibody, HRP conjugated may contribute to:
Understanding developmental disorders associated with Wnt pathway dysregulation
Identifying potential therapeutic targets in cancer, where Wnt signaling is often aberrantly activated
Elucidating basic molecular mechanisms of protein regulation and processing
Studying tissue-specific functions of TRABD2B, particularly in heart, kidney, and adipose tissues where it is enriched
TRABD2B (also known as Metalloprotease TIKI2) is a 517 amino acid metalloproteinase that functions as a negative regulator of Wnt signaling pathways. It mediates the cleavage of N-terminal residues in many Wnt proteins, thereby modulating their activity. Unlike its counterpart TIKI1 (which is required for head formation via Wnt cleavage-oxidation and inactivation), TIKI2 operates through specific metalloprotease activity that is dependent on Mn²⁺/Co²⁺ and can be inhibited by divalent metal chelators such as EDTA. In pathological contexts, TRABD2B has been found to be upregulated in renal cell carcinoma while appearing to suppress growth in osteosarcoma by targeting the canonical Wnt pathway .
The commercially available TRABD2B Antibody, HRP conjugated is a polyclonal antibody raised in rabbits using recombinant Human Metalloprotease TIKI2 protein (amino acids 195-405) as the immunogen. It is specifically reactive against human TRABD2B and has been validated for ELISA applications. The antibody is provided in liquid form with a storage buffer containing 0.03% Proclin 300 preservative, 50% glycerol, and 0.01M PBS at pH 7.4. It has been purified using antigen affinity chromatography and is of IgG isotype .
For optimal preservation of antibody activity, TRABD2B Antibody, HRP conjugated should be stored at -20°C or -80°C upon receipt. It is critical to avoid repeated freeze-thaw cycles as these can significantly compromise antibody function and sensitivity. For longer-term storage planning, the antibody has been demonstrated to maintain stability for up to 12 months from the date of receipt when stored at -20°C to -70°C. For projects requiring frequent access to the antibody, aliquoting is strongly recommended to minimize freeze-thaw cycles. When stored under sterile conditions after reconstitution, the antibody maintains stability for approximately 1 month at 2-8°C or 6 months at -20°C to -70°C .
When designing Western blot experiments using TRABD2B Antibody, HRP conjugated, several controls are essential for result validation:
Positive Control: Use lysates from HEK293 cells transfected with human TIKI2/TRABD2B. This provides confirmation of antibody specificity and expected band size (approximately 65 kDa under reducing conditions) .
Negative Control: Include mock-transfected HEK293 cells to demonstrate specificity and absence of non-specific binding .
Loading Control: Incorporate antibodies against housekeeping proteins such as GAPDH or β-actin to normalize protein loading.
Secondary Antibody Control: Run a lane with sample but without primary antibody to detect any non-specific binding of the secondary antibody.
Molecular Weight Marker: Include a protein ladder to accurately identify the TRABD2B band at approximately 65 kDa.
For optimal signal detection, a concentration of 0.5 μg/mL of the antibody is recommended, though this should be optimized for your specific experimental conditions .
For optimal utilization of TRABD2B Antibody, HRP conjugated in ELISA applications, follow these methodological considerations:
Coating Optimization: For direct ELISA, coat plates with purified TRABD2B protein (ideally using the recombinant human Metalloprotease TIKI2 protein spanning amino acids 195-405) at concentrations ranging from 0.1-1.0 μg/well.
Antibody Dilution Range: Begin with dilutions between 1:1000 and 1:5000 for the HRP-conjugated antibody. The optimal concentration should be determined empirically through a dilution series for each specific experimental setup .
Blocking Protocol: Use 2-5% BSA or non-fat milk in PBS to reduce background signal. Incubate for 1-2 hours at room temperature.
Detection System: Since the antibody is already HRP-conjugated, use a suitable substrate such as TMB (3,3',5,5'-Tetramethylbenzidine) for colorimetric detection or luminol-based reagents for enhanced chemiluminescence.
Standard Curve Preparation: Include a standard curve using recombinant TRABD2B protein at concentrations ranging from 0-1000 ng/mL to quantify unknown samples.
Remember that optimal dilutions and protocols should be determined by each laboratory for their specific application and experimental conditions .
To investigate TRABD2B's role in Wnt signaling, researchers can employ several complementary approaches:
Enzymatic Activity Assays: Assess TRABD2B's metalloprotease activity by incubating purified TRABD2B with Wnt proteins and analyzing cleavage products via mass spectrometry or Western blotting. Include controls with metalloprotease inhibitors like EDTA to confirm specificity .
TOPFlash Reporter Assays: Measure β-catenin-dependent transcriptional activity using TOPFlash luciferase reporter constructs in cells with modulated TRABD2B expression. This quantifies the functional impact of TRABD2B on canonical Wnt signaling output .
Co-immunoprecipitation Studies: Use anti-TRABD2B antibodies to pull down protein complexes and identify Wnt pathway components that physically interact with TRABD2B.
Cell-based Phenotypic Assays: In cell types where Wnt signaling drives specific phenotypes (e.g., osteoblast differentiation), assess how TRABD2B overexpression or knockdown affects these outcomes.
CRISPR-Cas9 Gene Editing: Generate TRABD2B knockout cell lines to comprehensively assess pathway changes in the absence of TRABD2B function.
These approaches can be combined with the use of HRP-conjugated TRABD2B antibodies for protein detection and quantification throughout experimental workflows.
While the HRP-conjugated version of the TRABD2B antibody is not optimal for flow cytometry due to the nature of the HRP conjugate, researchers can adapt protocols based on unconjugated TRABD2B antibody methodologies. Based on the available data with other TRABD2B antibody formats:
Cell Preparation: Harvest HEK293 cells transfected with human TIKI2/TRABD2B and untransfected controls. Wash cells in flow cytometry buffer (PBS with 2% FBS).
Antibody Adaptation: For the HRP-conjugated antibody, a modified protocol is required: first fix and permeabilize cells to allow intracellular staining if the target epitope is not surface-exposed.
Signal Development: After antibody incubation, cells must be washed thoroughly and incubated with a fluorogenic HRP substrate compatible with flow cytometry (such as PerCP, which produces fluorescence upon HRP activity).
Gating Strategy: Use forward and side scatter to identify the cell population of interest, and then analyze fluorescence intensity to quantify TRABD2B expression.
Controls: Include mock-transfected cells as negative controls to establish baseline fluorescence and determine positive staining thresholds .
For more standardized flow cytometry protocols, researchers might consider using specifically designed fluorochrome-conjugated antibodies rather than HRP-conjugated versions.
When investigating the relationship between TRABD2B expression and cancer progression using TRABD2B Antibody, HRP conjugated, researchers should consider several technical aspects:
Tissue Sample Preparation: For cancer tissue microarrays or patient-derived samples, optimize fixation conditions (typically 10% neutral buffered formalin) and antigen retrieval methods to ensure TRABD2B epitope accessibility.
Expression Quantification: Develop a standardized scoring system for immunohistochemistry to quantify TRABD2B expression levels across different cancer stages and grades. This can involve H-score methods (intensity × percentage of positive cells) or automated image analysis software.
Correlation Analysis: Design experiments to correlate TRABD2B expression levels with:
Functional Validation: Complement expression studies with functional assays in relevant cancer cell lines, manipulating TRABD2B levels and measuring effects on proliferation, migration, and invasion.
Technical Controls: For each experiment, include positive controls (tissues known to express TRABD2B) and negative controls (antibody diluent only) to validate staining specificity.
Optimizing dual immunofluorescence protocols using TRABD2B Antibody, HRP conjugated with other molecular markers requires careful consideration of several technical factors:
HRP Conversion System: Since the antibody is HRP-conjugated, researchers need to employ a tyramide signal amplification (TSA) system to convert HRP activity into fluorescence:
Use tyramide-conjugated fluorophores (e.g., tyramide-FITC or tyramide-Cy3)
The HRP catalyzes covalent binding of the fluorophore to proteins in the vicinity of the antibody
Sequential Staining Protocol:
Perform TRABD2B staining first using the HRP-conjugated antibody
Develop with appropriate tyramide-fluorophore
Thoroughly quench the HRP activity using 3% hydrogen peroxide
Proceed with the second primary antibody (from a different host species)
Detect using a directly conjugated fluorescent secondary antibody
Epitope Considerations:
When combining with Wnt pathway markers, prioritize antibodies targeting different regions of the signaling cascade
Consider detergent concentration and antigen retrieval methods that will work for both antigens
Spectral Compatibility:
Select fluorophores with minimal spectral overlap to avoid bleed-through
Include single-stained controls to confirm specificity and absence of cross-reactivity
Image Acquisition Parameters:
Use sequential scanning rather than simultaneous detection when using confocal microscopy
Establish exposure settings using control samples to prevent signal saturation
When encountering high background with TRABD2B Antibody, HRP conjugated, several common causes and solutions should be considered:
For particularly persistent background issues, consider implementing a stringent wash protocol using PBS-T (0.1% Tween-20) followed by high-salt washes (PBS with 0.5M NaCl) to disrupt non-specific ionic interactions .
When facing discrepancies between ELISA and Western blot results using TRABD2B Antibody, HRP conjugated, researchers should consider several factors in their analysis:
Epitope Accessibility Differences:
Post-translational Modifications:
Western blot may reveal multiple bands due to different glycosylation states or proteolytic processing
ELISA might preferentially detect certain modified forms based on epitope availability
Sample Preparation Considerations:
Cell lysis buffers for Western blot may preserve or disrupt certain protein interactions
Coating buffers for ELISA may partially denature proteins affecting antibody binding
Quantitative Analysis Approach:
Create a standardized calibration curve using recombinant TRABD2B protein
Normalize Western blot data to housekeeping proteins
Compare relative rather than absolute values between techniques
Validation Strategy:
Use alternative antibodies targeting different TRABD2B epitopes
Perform knockdown/overexpression experiments to confirm specificity
Consider mass spectrometry to identify proteins detected in conflicting results
When interpreting divergent results, consider that both methods provide complementary information: Western blot reveals size and potential modifications while ELISA provides more precise quantification under native conditions.
To address potential cross-reactivity of TRABD2B Antibody with other TRAB domain-containing proteins, researchers should implement the following experimental strategies:
Competitive Inhibition Assays:
Genetic Validation Approaches:
Test antibody reactivity in TRABD2B knockout cell lines generated via CRISPR-Cas9
Compare with TRABD2B-overexpressing cells as positive controls
Any persistent signal in knockout cells suggests cross-reactivity
Bioinformatic Analysis and Testing:
Identify sequence homology between TRABD2B and other TRAB domain proteins (particularly TRABD and TRABD2A)
Test antibody against recombinant proteins of these family members
Create a cross-reactivity profile detailing relative affinity for each protein
Immunoprecipitation-Mass Spectrometry:
Use the antibody for immunoprecipitation followed by mass spectrometry
Identify all proteins pulled down by the antibody
Quantify the relative abundance of TRABD2B versus other detected proteins
Epitope Mapping:
Generate a series of peptide fragments covering the immunogen region
Determine which specific sequences within the 195-405 AA region are recognized
Compare these epitopes with sequence alignments of related proteins
By systematically implementing these approaches, researchers can definitively characterize antibody specificity and account for any cross-reactivity in experimental interpretations.
When comparing TRABD2B Antibody, HRP conjugated with alternative detection systems for studying TIKI2's role in Wnt signaling modulation, several factors should be considered:
| Detection System | Advantages | Limitations | Best Applications |
|---|---|---|---|
| TRABD2B Antibody, HRP conjugated | Direct detection without secondary antibody; reduced protocol time; high sensitivity in ELISA applications | Limited fluorescence compatibility; potential higher background in some applications | ELISA, Western blot, IHC when signal amplification is needed |
| Unconjugated primary + HRP-secondary antibody | Signal amplification potential; greater flexibility in dilution optimization; broader application range | Longer protocol with additional incubation steps; potential for secondary cross-reactivity | Western blot, IHC, particularly when titration of detection sensitivity is required |
| Fluorophore-conjugated TRABD2B antibody | Direct visualization in microscopy; compatibility with multiplex staining; no substrate development needed | Potential photobleaching; may have lower sensitivity than enzymatic methods | Immunofluorescence, flow cytometry, live cell imaging |
| Genetic reporters (TRABD2B-GFP fusion) | Direct visualization of protein localization and dynamics; useful for live cell studies | Potential interference with native protein function; overexpression artifacts | Live-cell imaging, protein trafficking studies, FRAP experiments |
For comprehensive Wnt signaling studies, researchers should consider combining these approaches. For instance, use HRP-conjugated antibodies for quantitative analysis in ELISA and Western blot, while employing fluorescent approaches for subcellular localization studies. The specific experimental question should guide selection of the most appropriate detection system .
The methodological differences between sheep anti-human TRABD2B antibodies and rabbit polyclonal antibodies for TRABD2B detection involve several important technical considerations:
Host Species Considerations:
Epitope Recognition Patterns:
Application Optimization Requirements:
Sheep antibodies typically require higher concentrations (0.5 μg/mL recommended for Western blot)
Rabbit polyclonals often work at higher dilutions (1:1000-1:2000) for comparable applications
Signal Development Systems:
For the HRP-conjugated rabbit polyclonal, direct substrate addition is sufficient
Sheep antibodies require an additional secondary antibody incubation step with anti-sheep IgG-HRP
Cross-Reactivity Profiles:
Sheep antibodies may have different non-specific binding profiles than rabbit antibodies
This can be advantageous when working in tissues with high endogenous rabbit IgG
When selecting between these options, researchers should consider the specific experimental design, available detection systems, and potential for cross-reactivity with other proteins in their experimental system .
When encountering limited sensitivity with TRABD2B Antibody, HRP conjugated for detecting endogenous TRABD2B in specific cell types, researchers can implement several alternative approaches:
Signal Amplification Technologies:
Employ tyramide signal amplification (TSA) to enhance HRP-based detection
This catalytically deposits multiple tyramide-conjugated fluorophores or chromogens at the antibody binding site
Can increase sensitivity by 10-100 fold compared to conventional detection
Alternative Detection Platforms:
Switch to highly sensitive chemiluminescent substrates (SuperSignal West Femto)
Consider digital immunoassay platforms like Simoa or Single Molecule Counting that can detect proteins at femtomolar concentrations
Explore proximity ligation assay (PLA) which can detect single protein molecules through rolling circle amplification
Sample Enrichment Strategies:
Perform immunoprecipitation to concentrate TRABD2B before analysis
Use subcellular fractionation to isolate membrane fractions where TRABD2B is likely concentrated
Implement cell sorting to isolate subpopulations with higher TRABD2B expression
Transcriptional Analysis Complementation:
Combine protein detection with RT-qPCR to assess TRABD2B mRNA levels
Use RNA-scope in situ hybridization for sensitive detection of TRABD2B transcripts in tissue sections
Correlate transcript and protein data to validate low expression findings
Genetic Tagging Approaches:
For cultured cells, consider CRISPR knock-in of epitope tags (FLAG, HA, etc.) to the endogenous TRABD2B locus
Use well-characterized tag-specific antibodies with established high sensitivity
This maintains endogenous expression levels while improving detection sensitivity
By implementing these alternative strategies, researchers can overcome sensitivity limitations while still addressing their scientific questions regarding TRABD2B expression and function in challenging experimental systems .
To investigate TIKI2's metalloprotease activity on specific Wnt proteins using TRABD2B Antibody, HRP conjugated, researchers can implement the following experimental design:
In Vitro Cleavage Assay Development:
Express and purify recombinant Wnt proteins with N-terminal tags
Incubate with immunoprecipitated TRABD2B (using the antibody)
Analyze cleavage products via Western blot or mass spectrometry
Include controls with metalloprotease inhibitors (EDTA) and catalytically inactive TRABD2B mutants
Substrate Specificity Profiling:
Test multiple Wnt family members in parallel cleavage reactions
Quantify relative cleavage efficiency using densitometry of Western blots
Correlate cleavage patterns with Wnt protein sequence alignment to identify recognition motifs
Design synthetic peptide substrates based on identified cleavage sites for kinetic studies
Cell-Based Validation Approach:
Co-express TRABD2B and various Wnt proteins in HEK293 cells
Immunoprecipitate Wnt proteins from conditioned media and cell lysates
Analyze processing status using antibodies specific to N-terminal and C-terminal regions
Correlate processing with Wnt signaling activity using TOPFlash reporter assays
Kinetic Parameter Determination:
Design FRET-based peptide substrates containing identified TRABD2B cleavage sites
Measure reaction kinetics with purified TRABD2B enzyme
Determine Km and kcat values for different Wnt-derived substrates
Analyze the effect of divalent cations (Mn²⁺/Co²⁺) on enzyme activity
This comprehensive approach will provide mechanistic insights into how TRABD2B/TIKI2 regulates Wnt signaling through its metalloprotease activity .
When studying TRABD2B's role in cancer progression using tissue microarrays (TMAs) with TRABD2B Antibody, HRP conjugated, several methodological considerations are critical:
TMA Design and Sample Selection:
Include tissues representing different cancer stages and grades
Incorporate matched normal adjacent tissue controls for each patient
Consider inclusion of tissues known to have altered Wnt signaling
For renal cell carcinoma and osteosarcoma studies, include samples from different subtypes to account for molecular heterogeneity
Staining Protocol Optimization:
Perform antigen retrieval optimization (test pH 6.0 citrate and pH 9.0 EDTA buffers)
Titrate antibody concentration (start with 1:100 to 1:500 dilutions)
Use automated staining platforms when possible to ensure consistency
Include positive control tissues (based on TRABD2B expression data in Human Protein Atlas)
Scoring and Analysis Methodology:
Develop a standardized scoring system (e.g., H-score = intensity × percentage)
Employ digital pathology and automated image analysis for objective quantification
Have multiple pathologists score independently to establish inter-observer reliability
Correlate TRABD2B expression with established biomarkers and clinical outcomes
Validation Strategy:
Confirm TMA findings on whole tissue sections from a subset of cases
Validate protein expression with orthogonal methods (RNA-ISH, RT-qPCR from matched samples)
Perform statistical analysis accounting for multiple comparisons and potential confounding variables
Data Integration Approach:
Integrate TRABD2B expression data with genomic and clinical information
Perform multivariate analysis to assess independent prognostic value
Consider consensus molecular subtypes of the cancer type being studied
Analyze correlation with Wnt pathway activation markers
This comprehensive methodological approach will generate reliable data on TRABD2B's role in cancer progression that can be validated across patient cohorts .
To investigate the metal ion dependency of TRABD2B's enzymatic activity, researchers can design experiments utilizing TRABD2B Antibody, HRP conjugated in the following comprehensive approach:
Enzyme Purification and Activity Assay Development:
Immunoprecipitate TRABD2B from cell lysates using the antibody
Develop a fluorogenic or chromogenic peptide substrate based on known Wnt cleavage sites
Establish baseline activity measurements in standard buffer conditions
Metal Ion Dependency Profiling:
Systematically test enzyme activity across a panel of divalent cations:
Known cofactors: Mn²⁺, Co²⁺
Related metals: Zn²⁺, Fe²⁺, Mg²⁺, Ca²⁺, Ni²⁺, Cu²⁺
Create a concentration-response curve for each metal (0.1-10 mM)
Measure activity in terms of Vmax and Km for each condition
Chelation Studies:
Assess activity inhibition patterns with different chelators:
EDTA (broad spectrum)
EGTA (Ca²⁺-selective)
1,10-phenanthroline (Zn²⁺-selective)
Determine IC₅₀ values for each chelator
Test reversibility by metal ion replenishment after chelation
Structural Analysis Approaches:
Perform site-directed mutagenesis of predicted metal-coordinating residues
Measure activity of mutants in the presence of different metal ions
Use circular dichroism to assess structural changes in TRABD2B upon metal binding
Consider computational modeling of the metalloprotease domain
Cellular Validation:
Manipulate cellular metal ion availability using cell-permeable chelators
Monitor TRABD2B-dependent Wnt processing in these conditions
Correlate with functional Wnt signaling outputs (using TOPFlash assays)
Assess changes in TRABD2B subcellular localization in response to metal availability