WLS Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to WLS Antibody, Biotin Conjugated

WLS (Wntless Homolog, also known as GPR177) is a transmembrane protein critical for WNT signaling pathway regulation. The WLS Antibody, Biotin conjugated is a specialized immunoreagent designed for enhanced detection and amplification in assays such as ELISA, Western blotting (WB), and immunohistochemistry (IHC). Biotinylation enables binding to streptavidin or avidin conjugates, amplifying signal detection for low-abundance targets .

Biotin Conjugation Technology and Mechanism

Biotin conjugation involves chemically linking biotin molecules to the antibody’s Fc region or variable domains. This process preserves antibody specificity while enabling streptavidin-based detection systems.

Conjugation MethodKey FeaturesAdvantagesLimitations
ZBPA Domain ConjugationTargets Fc region via UV-mediated crosslinkingHigh specificity, minimal background staining Requires UV exposure, lower throughput
Lightning-Link KitsAmine-reactive labelingRapid (<20 mins), scalable (10 µg–100 mg) Potential nonspecific labeling of stabilizers (e.g., albumin)

Mechanism of Action:

  1. Primary Antibody Binding: The WLS antibody binds to its target antigen.

  2. Streptavidin-Avidin Interaction: Biotinylated antibodies bind to streptavidin-HRP, streptavidin-AP, or fluorescent streptavidin conjugates, enabling signal amplification .

Key Applications

ApplicationDetailsReferences
ELISAQuantitative detection of WLS in cell lysates or serum. Paired with streptavidin-HRP for colorimetric readouts .
Western BlottingDetection of WLS protein at ~52–63 kDa molecular weight. Requires HRP-conjugated streptavidin for enhanced sensitivity .
ImmunohistochemistryTissue localization of WLS using streptavidin-alkaline phosphatase or streptavidin-fluorophores .
Affinity PurificationIsolation of WLS protein complexes using streptavidin-agarose matrices .

Critical Studies

  1. ZBPA vs. Lightning-Link Conjugation:

    • ZBPA: Produces stringent labeling with no off-target staining, ideal for tissue microarrays .

    • Lightning-Link: Faster but may label stabilizers (e.g., albumin), causing nonspecific background .

  2. Signal Amplification Efficiency:

    • Biotinylated antibodies paired with streptavidin-HRP achieve >90% binding efficiency in pretargeting assays .

  3. Cross-Reactivity:

    • WLS antibodies (e.g., ABIN2462819) show reactivity with human, mouse, and dog species .

Comparison of Conjugation Methods for WLS Antibody

MethodConjugation TargetPurityThroughput
ZBPAFc region (UV-mediated)High (>90%)Low (labor-intensive)
Lightning-LinkPrimary aminesModerateHigh (scalable)

Challenges and Limitations

  • Endogenous Biotin Interference: Mitochondrial biotinylated proteins may cause background in IHC; blocking agents are required .

  • Concentration Sensitivity: Lightning-Link may fail at low antibody concentrations (<10 µg), necessitating buffer optimization .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery information.
Synonyms
WLS; C1orf139; GPR177; UNQ85/PRO18667; Protein wntless homolog; Integral membrane protein GPR177; Protein evenness interrupted homolog; EVI; Putative NF-kappa-B-activating protein 373
Target Names
WLS
Uniprot No.

Target Background

Function
WLS Antibody, Biotin conjugated, is involved in the regulation of Wnt protein sorting and secretion through a feedback mechanism. This interplay is critical in controlling the expression, subcellular localization, binding, and organelle-specific association of Wnt proteins. Furthermore, WLS plays a crucial role in establishing the anterior-posterior body axis during embryonic development.
Gene References Into Functions
  1. GPR177 has emerged as a potential prognostic marker and a promising therapeutic target for gastric cancer patients. PMID: 29555015
  2. Wls exhibits differential expression in Intrahepatic Cholangiocarcinoma tissues, with a positive correlation to tumor stage and lymphatic invasion. PMID: 29526886
  3. Research indicates that Wls expression varies across different stages of Hepatocellular Carcinoma (HCC), suggesting its potential role in HCC progression. PMID: 29127606
  4. The interaction between Wls and SEC12 is stable and characterized by a defined interface involving cytosolic segments of both proteins. Mutant Wls that fails to interact with the COPII machinery cannot effectively support Wnt secretion. This suggests that the formation of early Wnt secretory vesicles is tightly regulated to ensure the proper export of functional ligands. PMID: 28515233
  5. Studies have identified novel associations between WLS, ARHGAP1, and the 5' region of MEF2C, showing a strong correlation with bone mineral density compared to GWAS SNPs. PMID: 27616567
  6. Evidence suggests that Wls protein is associated with tumor metastasis and advanced TNM stage in gastric carcinoma, indicating its potential as a prognostic marker. PMID: 28110490
  7. Research indicates that WLS may play a significant role in the invasion and metastasis of colorectal carcinoma. PMID: 27102079
  8. Dysfunction of Wntless triggers the retrograde transport of Wingless from the Golgi to the ER, inducing ER stress. PMID: 26887613
  9. Genetic variations at the WLS and CCDC170/ESR1 loci have been found to be significantly associated with bone mineral density. PMID: 26911590
  10. A strong association between the expression of WLS and HER2 has been identified, which holds significant biological and clinical implications. PMID: 25258105
  11. This study identified CMTM8 as a potential tumor suppressor gene and GPR177 as a potential oncogene in osteosarcoma. PMID: 25551557
  12. GPR177 plays a crucial role in disease relapse and poor survival in patients with B-cell precursor acute lymphoblastic leukemia. PMID: 25115440
  13. Endogenous WLS interacts with Wnts in the endoplasmic reticulum, undergoes cycling to the plasma membrane, and subsequently returns to the endoplasmic reticulum via the Golgi apparatus. PMID: 24768165
  14. Colorectal tumors exhibit elevated levels of Wnt3 and GPR177. PMID: 24162018
  15. Data suggest that the common variants of WLS analyzed in this study are not associated with opioid or cocaine addiction. PMID: 23566366
  16. Evi expression in psoriatic skin biopsies is downregulated, suggesting that Evi-deficient mice develop skin lesions resembling human psoriasis. PMID: 23918954
  17. Findings suggest a mechanism by which Gpr177 regulates skeletal development through modulation of autocrine and paracrine Wnt signals in a lineage-specific manner. PMID: 23188710
  18. WLS functions as a negative regulator of melanoma proliferation and spontaneous metastasis by activating WNT/beta-catenin signaling. PMID: 23129487
  19. Overexpression of Evi/Wls is sufficient to promote downstream Wnt signaling. PMID: 22147553
  20. Research demonstrates that endocytosis motifs can be found in the intracellular loops of multipass membrane proteins. This study provides direct evidence that the trafficking of Wls is essential for efficient secretion of Wnt signaling proteins. PMID: 22027831
  21. The relatively ubiquitous expression of GPR177 suggests that this protein may regulate Wnt secretion in a variety of embryonic and adult tissue types. PMID: 20652957
  22. These findings suggest a novel role for GPR177 in regulating cellular responses to opioid agonist drugs, opening new avenues for investigating the pharmacotherapy of drug dependence. PMID: 20214800

Show More

Hide All

Database Links

HGNC: 30238

OMIM: 611514

KEGG: hsa:79971

STRING: 9606.ENSP00000346829

UniGene: Hs.647659

Protein Families
Wntless family
Subcellular Location
Golgi apparatus membrane; Multi-pass membrane protein. Cytoplasmic vesicle membrane; Multi-pass membrane protein. Cell membrane; Multi-pass membrane protein. Endoplasmic reticulum membrane; Multi-pass membrane protein. Golgi apparatus membrane; Multi-pass membrane protein. Early endosome membrane; Multi-pass membrane protein.

Q&A

What is the basic principle behind biotin-conjugated antibodies in immunological assays?

Biotin-conjugated antibodies function through the high-affinity interaction between biotin and streptavidin/avidin proteins. This interaction is one of the strongest non-covalent biological bonds (Kd ≈ 10^-15 M), making it extremely stable in various experimental conditions. In practical applications, the antibody is first labeled with multiple biotin molecules through chemical conjugation to reactive groups on the antibody (typically primary amines on lysine residues or sulfhydryl groups on reduced disulfides). This biotinylated antibody then binds to its target antigen, followed by detection using streptavidin or avidin conjugated to a reporter molecule (fluorophore, enzyme, etc.) . This system allows for signal amplification since multiple biotin molecules can be attached to a single antibody, and each biotin can recruit a streptavidin-reporter complex .

What are the standard applications for biotin-conjugated WLS antibodies?

Biotin-conjugated WLS antibodies are valuable tools in studying Wnt signaling pathways across multiple experimental platforms. The primary applications include:

ApplicationSample typeTypical dilutionDetection method
Western blottingCell/tissue lysates1:1000-1:6000Streptavidin-HRP
ELISAPurified proteins, serum1:1000-1:5000Streptavidin-HRP
ImmunohistochemistryTissue sections1:100-1:500Streptavidin-enzyme
ImmunofluorescenceFixed cells/tissues1:100-1:500Fluorophore-streptavidin
Flow cytometryCell suspensions1:50-1:200Fluorophore-streptavidin

While these applications are standard, it is recommended that researchers optimize the concentration of the biotin-conjugated antibody for each specific application and experimental system .

How should biotin-conjugated antibodies be stored to maintain activity?

Proper storage of biotin-conjugated antibodies is crucial for maintaining their activity and specificity. The recommended storage conditions are:

  • Store at -20°C for long-term storage

  • Avoid repeated freeze-thaw cycles by preparing small aliquots before freezing

  • For short-term storage (up to four weeks), 4°C is acceptable

  • Protect from exposure to light, particularly if the detection system includes fluorophores

  • Store in appropriate buffer conditions: typically PBS with 50% glycerol, 0.05% preservative, and 0.5% BSA at pH 7.2-7.4

  • Some antibodies are supplied in lyophilized form and should be reconstituted with deionized water immediately before use

Following these storage guidelines can significantly extend the shelf-life of the antibody, with most biotin-conjugated antibodies remaining stable for approximately one year from the date of receipt when properly stored .

What controls should be included when using biotin-conjugated WLS antibodies?

When designing experiments with biotin-conjugated WLS antibodies, the following controls are essential for reliable interpretation:

  • Isotype control: Use a biotin-conjugated antibody of the same isotype (e.g., IgG2a for mouse monoclonals) but irrelevant specificity to assess non-specific binding

  • Blocking control: Pre-incubate samples with unconjugated WLS antibody before adding biotin-conjugated WLS antibody to confirm binding specificity

  • No primary antibody control: Omit the biotin-conjugated WLS antibody but include all other reagents to assess background from the detection system

  • Positive control: Include a sample known to express WLS protein (based on literature or previous validation)

  • Negative control: Include a sample known not to express WLS protein or use WLS-knockout cells/tissues

  • Endogenous biotin blocking: Use an endogenous biotin-blocking kit, particularly for tissues known to contain high levels of endogenous biotin (e.g., liver, kidney)

Including these controls allows for accurate data interpretation and troubleshooting of any unexpected results.

How can biotin interference be mitigated in immunoassays using biotin-conjugated antibodies?

Biotin interference (BI) is a significant challenge in immunoassays using biotin-streptavidin technology, particularly with samples from subjects taking biotin supplements or receiving high-dose biotin therapy. Several strategies can effectively mitigate this interference:

  • Sample dilution adjustment: Increasing the minimum required dilution (MRD) can significantly reduce biotin interference. Studies have shown that adjusting the MRD from 10% to 1% can substantially decrease BI in bridge assays

  • Biotin depletion pretreatment: Implementing a streptavidin-based sample pretreatment step can effectively deplete free biotin from samples:

    • Incubate samples with streptavidin-coated paramagnetic beads

    • Allow 30-60 minutes for biotin binding

    • Magnetically separate and retain the supernatant

    • This approach has been shown to eliminate BI completely in various assay formats

  • Alternative detection technologies: Consider non-biotin-based detection systems for samples likely to contain high biotin levels:

    • Direct conjugation of enzymes or fluorophores to primary antibodies

    • Use of protein A/G-based detection systems

    • Implementation of directly labeled detection antibodies

  • Timing of sample collection: For research involving human subjects, collect samples before biotin supplementation or at least 8 hours after the last dose when possible

The appropriate mitigation strategy depends on the specific assay format, expected biotin levels, and required sensitivity. For critical applications, combining multiple approaches may be necessary .

What are the optimal methods for conjugating biotin to WLS antibodies while preserving epitope recognition?

Optimizing biotin conjugation to WLS antibodies requires balancing degree of biotinylation with antibody functionality. Several methods are available, each with distinct advantages:

  • NHS-ester chemistry (most common approach):

    • Uses N-hydroxysuccinimide (NHS) ester derivatives of biotin

    • Reacts with primary amines on lysine residues and the N-terminus

    • Optimal reaction conditions: pH 7.2-8.5, room temperature, 1-2 hours

    • Typically use a 5-20 molar excess of NHS-biotin

    • Purify by dialysis or gel filtration after reaction

  • Maleimide chemistry (for site-specific conjugation):

    • Targets reduced sulfhydryl groups in antibodies

    • Requires controlled reduction of disulfide bonds

    • More selective but technically challenging

    • Preserves antigen-binding regions more reliably

    • Reduction conditions: 2-10 mM DTT, 30 minutes at 37°C

  • Enzymatic methods (for controlled stoichiometry):

    • Use enzymes like transglutaminase for site-specific conjugation

    • Requires engineered antibodies with specific tag sequences

    • Provides precise control over biotinylation sites

    • Minimizes impact on antigen binding

The degree of biotinylation can be determined using HABA (4'-hydroxyazobenzene-2-carboxylic acid) assay, with optimal levels typically between 3-8 biotin molecules per antibody. Excessive biotinylation (>10 biotin molecules per antibody) can lead to decreased specificity and increased aggregation .

How can signal amplification systems be optimized when using biotin-conjugated WLS antibodies for detecting low-abundance proteins?

Detecting low-abundance WLS proteins requires sophisticated signal amplification strategies beyond standard detection methods:

  • Tyramide Signal Amplification (TSA):

    • Utilizes HRP-streptavidin to catalyze deposition of biotinylated tyramide

    • Creates multiple biotin molecules at the site of antibody binding

    • Can increase sensitivity 10-100 fold compared to conventional methods

    • Implementation protocol:
      a. Apply biotin-conjugated WLS antibody
      b. Add HRP-streptavidin conjugate
      c. Apply biotinylated tyramide substrate with H₂O₂
      d. Detect with fluorophore-labeled streptavidin

    • Example: The Biotin XX Tyramide SuperBoost Kit with streptavidin has been effectively used for detection of low-abundance proteins like ATP Synthase in HeLa cells

  • Multi-layer amplification:

    • Sequential application of biotinylated antibodies and streptavidin

    • Creates molecular layers that amplify signal

    • Protocol:
      a. Primary non-biotinylated WLS antibody
      b. Biotinylated secondary antibody
      c. Streptavidin (unconjugated)
      d. Biotinylated tertiary antibody
      e. Reporter-conjugated streptavidin

    • Can increase sensitivity 5-10 fold over conventional methods

  • Poly-HRP detection systems:

    • Uses streptavidin conjugated to polymers carrying multiple HRP molecules

    • Each biotin-binding site can recruit 10-20 HRP enzymes

    • Significantly increases sensitivity in chromogenic applications

    • Reduces incubation times while improving signal-to-noise ratio

Optimal method selection depends on the specific application, available instrumentation, and required sensitivity level. Titration experiments should be conducted to determine the optimal concentration of each reagent in the amplification system.

What troubleshooting approaches are recommended when biotin-conjugated WLS antibodies show non-specific binding?

Non-specific binding is a common challenge when using biotin-conjugated antibodies. The following systematic troubleshooting approach can help identify and resolve these issues:

  • Identify the source of non-specific binding:

    • Conduct parallel experiments with isotype controls to distinguish antibody-mediated from biotin-mediated non-specificity

    • Run samples on different cell/tissue types with known WLS expression profiles

    • Perform competition assays with unconjugated antibody to confirm specificity

  • Address endogenous biotin:

    • Implement an endogenous biotin blocking step using commercially available kits

    • Alternatively, use a streptavidin/avidin pretreatment followed by biotin blocking

    • This is particularly important for biotin-rich tissues such as liver, kidney, and brain

  • Optimize blocking conditions:

    • Test different blocking agents:

    Blocking AgentConcentrationAdvantagesLimitations
    BSA1-5%Widely compatibleSome biotin content
    Casein0.5-2%Low biotinLimited solubility
    Commercial blockersAs directedOptimized formulationsCost
    Normal serum5-10%Effective for IF/IHCSpecies considerations
    • Extend blocking time to 1-2 hours at room temperature

    • Consider adding 0.1-0.3% Triton X-100 to blocking buffer for membrane permeabilization

  • Adjust antibody concentration:

    • Titrate the biotin-conjugated antibody using 2-fold serial dilutions

    • Optimal concentration shows clear specific signal with minimal background

    • Typical working dilutions range from 1:100 to 1:6000 depending on application

  • Modify washing procedures:

    • Increase number of washes (5-6 times rather than the standard 3)

    • Extend washing time (5-10 minutes per wash)

    • Add 0.05-0.1% Tween-20 to wash buffers to reduce hydrophobic interactions

Implementation of these troubleshooting strategies should proceed systematically, changing one variable at a time and documenting results to identify the most effective approach for your specific experimental system.

How does the choice of linker between the antibody and biotin affect experimental outcomes?

The choice of linker between the antibody and biotin significantly impacts experimental performance across multiple dimensions:

The optimal linker choice depends on the specific application requirements, with longer water-soluble linkers (PEG-biotin) generally providing better performance in complex biological samples due to reduced steric hindrance and improved solubility .

What is the optimal protocol for using biotin-conjugated WLS antibodies in Western blotting?

The following optimized protocol ensures robust and specific detection of WLS protein using biotin-conjugated antibodies in Western blotting:

  • Sample preparation:

    • Lyse cells/tissues in RIPA buffer containing protease inhibitors

    • Determine protein concentration using BCA or Bradford assay

    • Prepare samples (20-50 μg total protein) in Laemmli buffer with reducing agent

    • Heat at 95°C for 5 minutes

  • Gel electrophoresis and transfer:

    • Separate proteins on 8-10% SDS-PAGE (WLS is approximately 62 kDa)

    • Transfer to PVDF membrane (0.45 μm pore size) at 100V for 1 hour or 30V overnight

    • Verify transfer efficiency with Ponceau S staining

  • Blocking and antibody incubation:

    • Block membrane with 5% non-fat milk in TBST for 1 hour at room temperature

    • For samples with high endogenous biotin, include a biotin blocking step using a commercial kit

    • Dilute biotin-conjugated WLS antibody to 1:1000-1:6000 in 1% BSA/TBST

    • Incubate membrane with antibody solution overnight at 4°C with gentle agitation

    • Wash 5× with TBST, 5 minutes each

  • Detection:

    • Incubate with HRP-conjugated streptavidin (1:5000-1:10000) in 1% BSA/TBST for 1 hour at room temperature

    • Alternatively, use fluorescently labeled streptavidin for multiplexing capabilities

    • Wash 5× with TBST, 5 minutes each

    • For chemiluminescent detection, apply ECL substrate and image using appropriate methods

    • For fluorescent detection, rinse with PBS and scan using appropriate wavelength settings

  • Controls and validation:

    • Include positive control (tissue/cell line known to express WLS)

    • Run parallel blot with isotype control antibody at equivalent concentration

    • For definitive specificity validation, include WLS knockout/knockdown samples

This protocol typically yields a distinct band at approximately 62 kDa corresponding to WLS protein, though observed molecular weight may vary slightly depending on post-translational modifications and experimental conditions .

How should researchers quantitatively assess the degree of biotinylation of WLS antibodies?

Accurate assessment of biotinylation levels is crucial for consistent experimental results. The following methods provide quantitative measurement of biotin incorporation:

  • HABA/Avidin Assay:

    • Based on the displacement of HABA (4'-hydroxyazobenzene-2-carboxylic acid) from avidin by biotin

    • Mix biotinylated antibody with HABA/avidin solution

    • Measure absorbance decrease at 500 nm

    • Calculate biotin:protein ratio using extinction coefficient (ε₅₀₀ = 34,000 M⁻¹cm⁻¹)

    • Quick and straightforward, but less sensitive than other methods

  • Fluorescent Biotin Quantification:

    • Incubate biotinylated antibody with fluorescently-labeled streptavidin

    • Measure fluorescence intensity after purification of complexes

    • Compare to standard curve of biotinylated protein standards

    • More sensitive than HABA assay, detecting as few as 1-2 biotin molecules per antibody

  • Mass Spectrometry:

    • Most precise method for determining biotinylation sites and stoichiometry

    • Digest biotinylated antibody with trypsin

    • Analyze peptides by LC-MS/MS

    • Identify biotinylated peptides by mass shift (+226 Da per biotin)

    • Provides site-specific information on biotinylation pattern

  • Dot Blot Approach:

    • Spot dilution series of biotinylated antibody on nitrocellulose

    • Probe with streptavidin-HRP

    • Compare signal intensity to biotinylated protein standards

    • Semi-quantitative but accessible for most laboratories

The optimal degree of biotinylation for WLS antibodies is typically 3-8 biotin molecules per antibody. Lower levels may provide insufficient sensitivity, while excessive biotinylation (>10 biotin molecules per antibody) may cause aggregation, increased non-specific binding, or compromised antigen recognition .

What strategies can minimize batch-to-batch variation when using biotin-conjugated antibodies in longitudinal studies?

Maintaining consistency across extended research timelines requires systematic approaches to minimize variability:

  • Standardized antibody characterization:

    • Determine biotin:protein ratio for each batch using HABA assay

    • Verify antigen binding using ELISA against recombinant WLS protein

    • Confirm specificity via Western blot against positive and negative controls

    • Document batch-specific performance metrics in standardized format

  • Reference standard implementation:

    • Create a large-scale "reference batch" of biotin-conjugated antibody

    • Aliquot and store at -80°C for long-term stability

    • Test each new batch against this reference standard

    • Establish acceptance criteria (e.g., within 20% of reference activity)

  • Calibration curve approach:

    • Develop a calibration curve using recombinant WLS protein

    • Run this curve with each experiment

    • Normalize experimental data to the calibration curve

    • This approach compensates for variations in detection sensitivity

  • Parallel sample processing:

    • When comparing samples from different timepoints, process them simultaneously

    • Use the same batch of all reagents including detection systems

    • Include internal control samples in each experimental run

    • Apply consistent image acquisition settings for all timepoints

  • Statistical approaches for post-hoc normalization:

    • Document batch information for all experiments

    • Apply statistical batch correction methods (e.g., ComBat, quantile normalization)

    • Include batch as a covariate in statistical analyses

    • Consider using mixed-effects models to account for batch effects

By implementing these strategies, researchers can significantly reduce technical variability while preserving the ability to detect true biological differences across longitudinal timepoints.

How can multiplex immunoassays be designed using biotin-conjugated WLS antibodies alongside other markers?

Designing effective multiplex assays with biotin-conjugated WLS antibodies requires careful consideration of several technical factors:

  • Compatible detection strategies:

    • Since one detection channel is occupied by the biotin-streptavidin system, other markers must use alternative detection methods:

    Primary detectionSecondary detection options for other markers
    Biotin-streptavidin-fluorophore 1Direct fluorophore conjugation
    Hapten systems (DNP, digoxigenin)
    Species-specific fluorescent secondaries
    Zenon labeling technology
  • Panel design considerations:

    • Ensure antibodies have compatible species origins to avoid cross-reactivity

    • Verify that detection fluorophores have minimal spectral overlap

    • Confirm that epitopes are accessible in the fixation/permeabilization conditions required

    • Example 4-color panel for Wnt signaling analysis:

    TargetPrimary AbDetection systemFluorophore
    WLSBiotin-conjugated anti-WLSStreptavidinAlexa Fluor 488
    β-cateninRabbit anti-β-cateninAnti-rabbitAlexa Fluor 555
    LRP6Goat anti-LRP6Anti-goatAlexa Fluor 647
    DAPIN/ADirect bindingDAPI/BFP
  • Optimization protocol:

    • Test each antibody individually before combining

    • Titrate each primary antibody to determine optimal concentration

    • Perform blocking of endogenous biotin if tissue samples are used

    • Include fluorescence-minus-one (FMO) controls for each marker

    • Validate multiplexed staining against single-marker controls

  • Image acquisition considerations:

    • Capture images sequentially rather than simultaneously to minimize bleed-through

    • Include single-stained controls for spectral unmixing if necessary

    • Maintain consistent exposure settings across experimental samples

    • Apply appropriate background subtraction methods

  • Data analysis approaches:

    • Measure co-localization using Pearson's or Mander's coefficients

    • Consider automated segmentation for quantitative analysis

    • Apply batch correction if analyzing images from multiple experiments

This approach enables comprehensive analysis of Wnt signaling pathway components while maintaining specificity and quantitative accuracy.

How should researchers design validation experiments for newly acquired biotin-conjugated WLS antibodies?

Comprehensive validation is essential before using biotin-conjugated WLS antibodies in critical experiments. The following systematic validation framework ensures antibody reliability:

  • Specificity validation:

    • Genetic approach: Test antibody against WLS knockout and wildtype samples

    • Molecular approach: Test against cells with WLS overexpression

    • Peptide competition: Pre-incubate antibody with immunizing peptide to block specific binding

    • Cross-species reactivity: Test predicted reactive species to confirm specificity

  • Technical validation across applications:

    ApplicationValidation criteriaEssential controls
    Western blotSingle band at expected MWLysate from KO cell line
    IHC/IFExpected subcellular patternSecondary-only control
    IPEnrichment of target proteinIgG control
    Flow cytometryPopulation shift vs. isotypeBlocking experiment
  • Sensitivity assessment:

    • Determine limit of detection using serial dilutions of recombinant WLS protein

    • Compare sensitivity to non-biotinylated version of the same antibody

    • Assess signal-to-noise ratio across various sample types

  • Reproducibility testing:

    • Repeat key experiments on different days

    • Test multiple antibody dilutions to identify robust working range

    • If possible, compare results from different lots of the same antibody

  • Application-specific optimization:

    • For each intended application, determine:

      • Optimal antibody concentration

      • Incubation conditions (time, temperature)

      • Sample preparation requirements

      • Detection system parameters

  • Documentation:

    • Create detailed validation report including:

      • Images of positive and negative controls

      • Quantitative assessment of signal-to-noise ratio

      • Optimal working conditions for each application

      • Batch/lot information for future reference

This systematic validation approach provides confidence in antibody performance and facilitates troubleshooting if unexpected results occur in subsequent experiments.

What statistical approaches are recommended for analyzing data from experiments using biotin-conjugated WLS antibodies?

  • Quantification normalization strategies:

    • For Western blot: Normalize WLS signal to loading controls (GAPDH, β-actin, total protein)

    • For IHC/IF: Use ratio to DAPI or other cellular compartment marker

    • For flow cytometry: Report median fluorescence intensity (MFI) or percent positive

  • Addressing non-normal distributions:

    • Test data for normality using Shapiro-Wilk or Kolmogorov-Smirnov tests

    • For non-normal data, apply log or other appropriate transformations

    • If transformation is ineffective, use non-parametric tests:

    Parametric testNon-parametric alternative
    t-testMann-Whitney U test
    ANOVAKruskal-Wallis test
    Pearson correlationSpearman correlation
  • Accounting for technical variability:

    • Include technical replicates (minimum 3) for each biological sample

    • Use nested analysis approaches to separate technical from biological variation

  • Multiple testing correction:

    • When analyzing multiple experimental conditions or timepoints

    • Apply Benjamini-Hochberg procedure for false discovery rate (FDR) control

    • Use Bonferroni correction when strict family-wise error rate (FWER) control is needed

    • Report both raw and adjusted p-values

  • Effect size reporting:

    • Go beyond p-values to report effect sizes (Cohen's d, fold change)

    • Include confidence intervals for all reported effects

    • Consider minimum effect size of biological significance

  • Power analysis for experimental design:

    • Use preliminary data to estimate variance

    • Determine sample size needed for detecting biologically meaningful differences

These statistical approaches ensure robust analysis and interpretation of WLS expression data while accounting for the technical characteristics of biotin-conjugated antibody detection systems.

How can researchers effectively troubleshoot when biotin-conjugated WLS antibodies generate contradictory results across different experimental platforms?

Contradictory results across platforms require systematic investigation to identify and resolve underlying issues:

  • Establish a comparison framework:

    • Document all experimental variables across platforms:

      • Sample preparation methods

      • Buffer compositions

      • Incubation conditions

      • Detection systems

    • Create a decision tree for systematic evaluation

  • Epitope accessibility assessment:

    • Different applications expose different epitopes:

      • Western blot: Denatured epitopes

      • IHC/IF: Fixed/crosslinked epitopes

      • Flow cytometry: Native cell surface epitopes

    • Test alternative fixation/permeabilization methods

    • Consider epitope retrieval approaches for IHC

  • Sample-specific interference evaluation:

    • Test for endogenous biotin interference using streptavidin-only controls

    • Assess matrix effects by spike-in recovery experiments

    • Check for post-translational modifications that may affect epitope recognition

    • Implement specific blocking steps for problematic samples:

    Interference sourceBlocking strategy
    Endogenous biotinAvidin/biotin blocking kit
    Non-specific bindingExtended serum block (5-10%)
    Fc receptor bindingFc receptor blocking reagent
    Cross-reactivityPre-absorption with related proteins
  • Antibody validation reconciliation:

    • Perform side-by-side comparison with alternative WLS antibodies

    • Implement orthogonal detection methods (e.g., mass spectrometry)

    • Correlate results with functional readouts of Wnt signaling

    • Consider genetic approaches (siRNA, CRISPR) to validate specificity

  • Technical optimization:

    • Systematically titrate antibody concentration for each platform

    • Test multiple detection systems (different streptavidin conjugates)

    • Optimize signal-to-noise ratio through washing and blocking adjustments

    • Consider signal amplification systems for low-abundance detection

  • Biological interpretation framework:

    • Recognize that different platforms may detect different aspects of biology

    • Western blot quantifies total protein regardless of localization

    • IHC/IF reveals spatial distribution but may be less quantitative

    • Flow cytometry measures only accessible epitopes

    • Integrate results across platforms for comprehensive understanding

By systematically addressing these factors, researchers can resolve contradictions and develop a coherent understanding of WLS expression and function across experimental systems.

How can biotin-conjugated WLS antibodies be effectively utilized in single-cell analysis techniques?

Single-cell analysis with biotin-conjugated WLS antibodies enables unprecedented insights into heterogeneous cell populations:

  • Single-cell mass cytometry (CyTOF) integration:

    • Utilize biotinylated WLS antibody with isotope-labeled streptavidin

    • Optimal metal tags: 153Eu, 154Sm, or 174Yb streptavidin conjugates

    • Include barcoding strategy for batch analysis

    • Implementation protocol:
      a. Fix cells in 1.6% paraformaldehyde
      b. Permeabilize with methanol if intracellular staining is required
      c. Block with 2% BSA in PBS
      d. Incubate with biotin-conjugated WLS antibody (1:100)
      e. Wash 3× with PBS/0.5% BSA
      f. Add metal-labeled streptavidin (1:100)
      g. Wash 3× before acquisition

  • Imaging mass cytometry applications:

    • Apply biotinylated WLS antibody to tissue sections

    • Detect with metal-tagged streptavidin

    • Enables multiplexing with 40+ additional markers

    • Preserves spatial context of WLS expression

  • Single-cell RNA-protein correlation:

    • CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing):
      a. Conjugate oligonucleotide barcodes to streptavidin
      b. Apply biotin-WLS antibody to live cells
      c. Detect with oligo-tagged streptavidin
      d. Sequence both transcriptome and protein tags

    • Enables direct correlation between WLS protein levels and gene expression

  • Microfluidic approaches:

    • Single-cell Western blotting with biotin-conjugated antibodies

    • Droplet-based antibody detection systems

    • Microfluidic image cytometry with streptavidin-fluorophore detection

  • Advanced computational analysis:

    • Dimensionality reduction techniques (tSNE, UMAP) to visualize WLS expression patterns

    • Trajectory inference to map WLS expression changes during differentiation

    • Clustering approaches to identify WLS-expressing subpopulations

    • Integration with other single-cell omic datasets

These approaches enable comprehensive understanding of WLS expression heterogeneity at single-cell resolution, providing insights into Wnt signaling dynamics across diverse cell types.

What recent technological advances have improved the specificity and sensitivity of biotin-conjugated antibody applications?

Recent technological innovations have significantly enhanced the performance of biotin-conjugated antibodies:

  • Enhanced conjugation chemistries:

    • Site-specific conjugation using engineered antibodies:

      • THIOMAB technology: Engineered cysteine residues for site-specific labeling

      • Sortase-mediated conjugation: Enzymatic labeling at specific C-terminal sequences

      • Transglutaminase-based approaches: Q-tag systems for controlled biotinylation

    • These approaches maintain native antigen binding while providing consistent biotin positioning

  • Next-generation signal amplification:

    • Digital counting technologies:

      • Single-molecule array (Simoa) platforms

      • Digital ELISA formats with femtomolar sensitivity

    • Branched DNA amplification:

      • RNAscope-like technologies adapted for protein detection

      • Can improve sensitivity by 100-1000 fold over conventional detection

  • Advanced detection systems:

    • Quantum dot-streptavidin conjugates:

      • Enhanced brightness and photostability

      • Narrow emission spectra for improved multiplexing

    • Lanthanide-based time-resolved fluorescence:

      • Elimination of autofluorescence through time-gated detection

      • Improved signal-to-noise in challenging tissue types

  • Biotin mimetics and alternatives:

    • Development of biotin analogs with reduced endogenous interference:

      • Desthiobiotin: Lower affinity but reversible binding

      • Iminobiotin: pH-dependent binding for controlled elution

    • Alternative tag systems:

      • Click chemistry approaches (DBCO, tetrazine ligation)

      • HaloTag and SNAP-tag technologies

  • AI-enhanced image analysis:

    • Deep learning algorithms for automated quantification:

      • Convolutional neural networks for pattern recognition

      • Instance segmentation for single-cell quantification

    • Improved sensitivity through computational clearing:

      • Deconvolution algorithms

      • Background subtraction neural networks

These technological advances collectively enable more precise, sensitive, and quantitative analysis of WLS expression across diverse experimental platforms, supporting deeper insights into Wnt signaling biology.

What are the key considerations for choosing between biotin-conjugated antibodies and alternative detection systems for WLS protein studies?

Selecting the optimal detection system for WLS protein analysis requires balancing multiple factors to align with specific research objectives:

  • Sensitivity requirements:

    • Biotin-streptavidin systems offer signal amplification advantages for detecting low-abundance WLS protein

    • Alternative direct conjugation systems may provide sufficient sensitivity for high-expression contexts

    • Consider the biological context: WLS expression varies significantly across tissue types and cellular states

  • Sample type constraints:

    • Tissues with high endogenous biotin (liver, kidney, brain) may require alternative detection systems

    • Clinical samples from patients on biotin supplementation may show interference

    • Fresh vs. fixed tissues have different optimal detection approaches

  • Multiplexing needs:

    • When extensive multiplexing is required, direct fluorophore conjugation may be preferable

    • For co-localization studies with 2-3 targets, biotin-streptavidin systems work well with complementary detection methods

    • Consider spectral overlap and cross-reactivity in multiplexed systems

  • Quantification precision:

    • Direct conjugates provide more linear signal-to-concentration relationships

    • Biotin-amplified signals offer enhanced sensitivity but potentially reduced linearity at high concentrations

    • Alternative systems like mass cytometry provide highly quantitative data independent of fluorescence

  • Experimental workflow considerations:

    • Biotin systems add additional incubation and washing steps

    • Direct conjugates streamline protocols but may require higher antibody concentrations

    • Consider time constraints and throughput requirements

  • Cost-benefit analysis:

    • Biotin conjugation is relatively inexpensive and widely available

    • Specialized conjugation methods may offer superior performance but at higher cost

    • Consider long-term experimental needs and sample availability

The optimal detection strategy should be selected based on the specific research question, experimental constraints, and performance requirements. For many applications, biotin-conjugated WLS antibodies offer an excellent balance of sensitivity, specificity, and flexibility, particularly when combined with appropriate controls and optimization.

What emerging research directions are likely to benefit from the application of biotin-conjugated WLS antibodies?

The unique properties of biotin-conjugated WLS antibodies position them as valuable tools for several cutting-edge research areas:

  • Spatial transcriptomics integration:

    • Combining in situ hybridization with protein detection

    • Correlating WLS protein localization with transcriptional profiles

    • Mapping protein-RNA relationships in tissue microenvironments

    • Will provide unprecedented insights into post-transcriptional regulation of Wnt signaling

  • Extracellular vesicle characterization:

    • Detection of WLS in exosomes and microvesicles

    • Flow cytometric analysis of WLS+ vesicle populations

    • Super-resolution microscopy of vesicle-associated WLS

    • May reveal novel intercellular communication mechanisms in development and disease

  • Organoid technology applications:

    • Tracking WLS expression during organoid development

    • Isolation of WLS-expressing stem/progenitor populations

    • Live imaging of Wnt pathway dynamics

    • Will enhance understanding of tissue morphogenesis and self-organization

  • Drug discovery applications:

    • High-content screening for WLS modulators

    • Target engagement studies for Wnt pathway inhibitors

    • Patient-derived xenograft analysis

    • Could identify novel therapeutic approaches for Wnt-dependent cancers

  • Clinical biomarker development:

    • Tissue microarray analysis of WLS expression

    • Circulating tumor cell detection

    • Minimally invasive diagnostic approaches

    • May improve prognostic assessment and treatment stratification

  • Regenerative medicine applications:

    • Monitoring WLS in stem cell differentiation protocols

    • Quality control for cell therapy products

    • Tracking transplanted cell populations

    • Could enhance development of cell-based therapies

These emerging research directions highlight the continued value of biotin-conjugated WLS antibodies in advancing our understanding of Wnt signaling biology and its translational applications. As detection technologies continue to evolve, these antibodies will remain important tools in the scientific toolkit, particularly for applications requiring high sensitivity and specificity.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.