WWC3 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically ship orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery estimates, please contact your local distributor.
Synonyms
WWC3 antibody; KIAA1280 antibody; Protein WWC3 antibody
Target Names
WWC3
Uniprot No.

Q&A

What is FITC conjugation and how does it work with antibodies?

Fluorescein isothiocyanate (FITC) is one of the most commonly used fluorescent dyes for antibody labeling in fluorescence-activated cell sorting (FACS) analysis and immunofluorescence applications. FITC is a small organic molecule that conjugates to proteins via primary amines, particularly lysines present in antibodies. The conjugation reaction involves the isothiocyanate reactive group (-N=C=S) on FITC forming stable thiourea bonds with primary amines on the antibody .

The process creates a covalent attachment that allows the antibody to retain its binding specificity while gaining fluorescent properties. FITC-conjugated antibodies typically contain between 3 and 6 FITC molecules per antibody molecule. This conjugation ratio is carefully controlled because higher conjugation levels can result in solubility problems and internal quenching that reduces fluorescence intensity .

What are the spectral properties of FITC-conjugated antibodies?

FITC-conjugated antibodies have specific excitation and emission properties that determine their compatibility with various imaging instruments and experimental designs. FITC is typically excited by the 488 nm line of an argon laser, and its emission is collected at approximately 530 nm . These spectral characteristics make FITC-conjugated antibodies compatible with standard fluorescence microscopes, flow cytometers, and imaging systems equipped with appropriate filter sets.

It's important to note that FITC is available as a mixture of isomers, primarily fluorescein 5-isothiocyanate (5-FITC) and fluorescein 6-isothiocyanate (6-FITC), which have slightly different spectral properties . This mixture is identified by the CAS number 27072-45-3, while the individual isomers have their own identifiers (3326-32-7 for 5-isomer and 18861-78-4 for 6-isomer) .

What applications are most suitable for FITC-conjugated WWC3 antibodies?

FITC-conjugated antibodies are widely used in various immunological techniques. Based on information from similar antibody conjugates, FITC-conjugated WWC3 antibodies would be applicable for:

  • Western Blotting (WB): Used at dilutions ranging from 1:300 to 1:10000, depending on the specific antibody preparation and sample type .

  • Immunofluorescence (IF): Both for paraffin-embedded tissue sections (IHC-P) and cell cultures (ICC), typically used at dilutions of 1:50-200 .

  • Flow Cytometry: For quantitative analysis of WWC3 expression in cell populations.

  • ELISA: For quantitative detection of WWC3 in solution samples .

The versatility of FITC conjugation makes these antibodies valuable tools for multiple experimental approaches within the same research project.

How can I optimize the FITC conjugation ratio for maximum sensitivity without compromising antibody function?

Optimizing the FITC:antibody ratio is crucial for achieving maximum sensitivity while maintaining antibody functionality. The ideal conjugation typically ranges between 3-6 FITC molecules per antibody, but this should be empirically determined for each specific antibody .

Methodological approach:

  • Perform parallel conjugation reactions with different molar ratios of FITC to antibody (e.g., 10:1, 20:1, and 30:1).

  • Purify each conjugate using desalting columns to remove unreacted FITC.

  • Determine the degree of labeling (DOL) by measuring absorbances at 280 nm (protein) and 495 nm (FITC) using the formula:
    DOL=A495×MWantibodyεFITC×[antibody]DOL = \frac{A_{495} \times MW_{antibody}}{\varepsilon_{FITC} \times [antibody]}
    Where ε₍FITC₎ is the extinction coefficient of FITC (approximately 70,000 M⁻¹cm⁻¹)

  • Test each conjugate for:

    • Brightness in flow cytometry or microscopy

    • Background staining/non-specific binding

    • Antigen recognition (comparing to unconjugated antibody)

  • Select the conjugate that offers the optimal balance between fluorescence intensity and maintained antibody specificity.

What are the most effective strategies for troubleshooting low signal intensity with FITC-conjugated WWC3 antibodies?

When encountering low signal intensity with FITC-conjugated antibodies, several methodological approaches can help identify and resolve the issue:

  • Check for photobleaching: FITC is relatively susceptible to photobleaching. Minimize exposure to light during storage and preparation. Use anti-fade mounting media for microscopy samples and consider adding anti-photobleaching agents.

  • Verify pH conditions: FITC fluorescence is pH-dependent, with optimal emission at slightly alkaline pH (7.5-8.5). Ensure buffers are at appropriate pH and consider using buffered mounting media.

  • Optimize antibody concentration: Titrate the antibody to determine the optimal working concentration. Too low concentrations result in weak signals, while too high concentrations may increase background.

  • Signal amplification: For samples with low target expression, consider using:

    • Tyramide signal amplification (TSA)

    • Secondary anti-FITC antibodies conjugated to brighter fluorophores

    • Biotin-streptavidin systems

  • Evaluate quenching factors: High protein concentrations or certain buffer components can quench fluorescence. Modify sample preparation protocols accordingly.

  • Check instrument settings: Ensure flow cytometer or microscope settings are optimized for FITC detection, including appropriate laser power, filter sets, and detector sensitivity.

  • Consider alternative conjugates: If signal remains problematic, consider switching to a brighter fluorophore like Alexa Fluor 488, which has similar spectral properties but greater photostability.

How does the storage of FITC-conjugated antibodies affect their performance, and what are the best preservation methods?

Proper storage is critical for maintaining the performance of FITC-conjugated antibodies. The fluorophore and protein components can both degrade under suboptimal conditions.

Storage recommendations and performance implications:

Storage ConditionStability PeriodEffect on PerformanceNotes
-20°C to -70°C6-12 monthsMaintains activity and fluorescenceRecommended for long-term storage
2-8°C (refrigerated)1 monthGradual loss of fluorescence intensityAcceptable for short-term use
Room temperatureHours to daysRapid fluorescence loss and protein degradationAvoid except during experiment execution

Best preservation methods:

  • Buffer composition: Store in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 to maintain stability .

  • Aliquoting: Divide into small single-use aliquots to avoid repeated freeze-thaw cycles, which significantly reduce antibody performance .

  • Light protection: Store in amber or opaque containers to protect from light exposure, which accelerates photobleaching.

  • Additives: Addition of protein stabilizers like 1% BSA can help maintain antibody integrity .

  • Temperature stability: Maintain consistent storage temperature; temperature fluctuations can accelerate degradation.

Performance should be validated after extended storage periods by testing against fresh conjugates or positive controls to ensure sensitivity and specificity are maintained.

What controls should be included when using FITC-conjugated WWC3 antibodies in fluorescence microscopy or flow cytometry?

Proper experimental controls are essential for generating reliable and interpretable data with FITC-conjugated antibodies. The following controls should be incorporated into experimental designs:

Essential controls for fluorescence microscopy and flow cytometry:

  • Isotype control: Use a FITC-conjugated antibody of the same isotype (e.g., IgG) but with irrelevant specificity to assess non-specific binding and establish background threshold levels.

  • Unstained sample: Include samples without any antibody to determine autofluorescence levels of the cells or tissues being studied.

  • Secondary antibody control (if applicable): When using indirect detection methods, include samples treated only with the secondary antibody to assess non-specific binding.

  • Blocking validation: Include samples where the primary binding site is blocked with unconjugated antibody or recombinant antigen to confirm specificity.

  • Positive control: Include samples known to express WWC3 to validate that the staining protocol is working correctly.

  • Negative control: Include samples known not to express WWC3 to confirm specificity.

  • Compensation controls (for multicolor flow cytometry): Single-color controls to compensate for spectral overlap between fluorophores.

These controls help distinguish true positive signals from artifacts and provide confidence in interpreting experimental results.

How can I minimize background fluorescence when working with FITC-conjugated antibodies in tissue sections?

Background fluorescence can significantly impact the signal-to-noise ratio when using FITC-conjugated antibodies, particularly in tissue sections that may contain autofluorescent components. Several methodological approaches can reduce background:

  • Optimize fixation: Overfixation with aldehydes can increase autofluorescence. Use the minimum fixation time necessary and consider using alternative fixatives like zinc-based fixatives that produce less autofluorescence.

  • Effective blocking: Use a combination of serum (5-10%) from the species in which the secondary antibody was raised, plus 1-3% BSA to block non-specific binding sites.

  • Autofluorescence quenching:

    • For formalin-fixed tissues: Pretreat with 0.1-1% sodium borohydride

    • For lipofuscin: Use Sudan Black B (0.1-0.3%)

    • For general autofluorescence: Incubate with 0.1% Toluidine Blue or 1mM CuSO₄ in 50mM ammonium acetate buffer

  • Optimize antibody concentration: Titrate the FITC-conjugated antibody to find the optimal concentration that maximizes specific signal while minimizing background.

  • Washing optimization: Increase the number, duration, and volume of washes with 0.05-0.1% Tween-20 in PBS to remove unbound antibody.

  • Counterstain selection: Choose counterstains that don't interfere with FITC fluorescence (avoid propidium iodide which has spectral overlap).

  • Confocal microscopy settings: Adjust pinhole settings to minimize out-of-focus fluorescence that contributes to background.

What are the advantages and limitations of FITC compared to other fluorophores for antibody conjugation in multicolor immunofluorescence?

Understanding the comparative properties of FITC and other fluorophores is crucial for designing optimal multicolor immunofluorescence experiments:

Advantages of FITC:

  • Well-established fluorophore with standardized protocols and wide availability .

  • Compatible with most common fluorescence detection systems and filter sets.

  • Relatively small molecule that typically doesn't interfere with antibody binding .

  • Economical compared to newer proprietary fluorophores.

  • Can be detected by anti-FITC antibodies for signal amplification .

Limitations of FITC:

  • Relatively rapid photobleaching compared to newer fluorophores like Alexa Fluors.

  • pH sensitivity - fluorescence decreases significantly at acidic pH.

  • Moderate quantum yield compared to some newer fluorophores.

  • Broader emission spectrum that can create spectral overlap in multicolor experiments.

  • Susceptible to quenching at higher conjugation ratios .

Comparison with alternative fluorophores for multicolor applications:

FluorophoreExcitation/Emission (nm)Relative BrightnessPhotostabilitypH SensitivityCost
FITC495/519ModerateLowHighLow
Alexa Fluor 488496/519HighHighLowHigh
PE (Phycoerythrin)565/575Very HighModerateLowHigh
Cy3550/570HighModerateLowModerate
Dylight 488493/518HighHighLowModerate

For multicolor immunofluorescence, consider using Alexa Fluor 488 instead of FITC when targeting low-abundance proteins like WWC3, as it provides better photostability and brightness. Reserve FITC for abundant targets or when cost is a significant factor.

How can I quantitatively assess the degree of labeling (DOL) of my FITC-conjugated WWC3 antibodies?

Determining the degree of labeling (DOL) is crucial for ensuring consistent experimental results and proper interpretation of fluorescence intensity data. The following methodological approach provides a reliable quantitative assessment:

Spectrophotometric method for DOL determination:

  • Measure the absorbance of the purified FITC-antibody conjugate at both 280 nm (A₂₈₀, protein absorption) and 495 nm (A₄₉₅, FITC absorption).

  • Calculate the protein concentration using the corrected A₂₈₀:
    A280(corrected)=A280(A495×0.35)A_{280(corrected)} = A_{280} - (A_{495} \times 0.35)
    Where 0.35 is the correction factor for FITC's contribution to absorption at 280 nm.

  • Calculate the FITC concentration using A₄₉₅:

[FITC] = \frac{A_{495}}{\varepsilon_{FITC}} $$
Where ε₍FITC₎ is the extinction coefficient of FITC (approximately 70,000 M⁻¹cm⁻¹)

  • Calculate the DOL:
    DOL=[FITC][protein]DOL = \frac{[FITC]}{[protein]}

Interpretation guidelines:

DOL RangeInterpretationRecommended Applications
0.5-2Low labelingGood for highly expressed targets, minimal risk of functional impact
3-6Optimal labelingBalanced brightness and functionality for most applications
7-10High labelingMaximum brightness but potential functional impact, good for low-abundance targets
>10Excessive labelingPotential quenching and solubility issues, not recommended

For WWC3 antibodies specifically, aim for a DOL in the 3-6 range for most research applications, as this provides sufficient fluorescence while minimizing the risk of altered binding properties or solubility issues.

What are the best approaches for analyzing FITC signal intensity in quantitative studies of WWC3 expression?

Quantitative analysis of FITC signal intensity requires careful attention to standardization, calibration, and normalization procedures to ensure reliable and reproducible results:

Flow cytometry analysis approaches:

  • Median Fluorescence Intensity (MFI): Preferred over mean fluorescence intensity as it is less affected by outliers.

  • Standard beads calibration: Use calibrated FITC beads to convert arbitrary fluorescence units to Molecules of Equivalent Soluble Fluorochrome (MESF) for absolute quantification.

  • Normalization strategies:

    • Subtract autofluorescence (from unstained samples)

    • Calculate the ratio of sample MFI to isotype control MFI

    • Use the formula: (Sample MFI - Isotype MFI) / Isotype MFI

Microscopy-based quantification approaches:

  • Integrated density measurement: Calculate the product of area and mean gray value to account for both signal intensity and size of positive regions.

  • Background subtraction: Use adjacent negative regions or isotype controls to determine and subtract background levels.

  • Colocalization analysis: For double-labeling experiments, calculate Pearson's correlation coefficient or Mander's overlap coefficient between WWC3 and other markers.

  • Z-stack considerations: When analyzing 3D samples, use maximum intensity projections or sum slices appropriately, ensuring consistent approach across all samples.

Software tools for quantification:

  • ImageJ/FIJI with appropriate plugins

  • CellProfiler for automated cell-based analysis

  • FlowJo or FCS Express for flow cytometry data

  • QuPath for tissue section analysis

For all quantitative studies, include appropriate positive and negative controls, and report both raw and normalized values for transparency.

How can I distinguish between specific WWC3 signal and non-specific binding or autofluorescence in challenging tissue samples?

Differentiating specific signal from non-specific binding or autofluorescence is crucial for accurate interpretation of results, particularly in tissues known to have high autofluorescence like liver, brain, and aged tissues:

Advanced methodological approaches:

  • Spectral unmixing: Use spectral detectors (available on some confocal microscopes) to separate FITC signal from autofluorescence based on their distinct spectral signatures.

  • Lifetime imaging: Fluorescence Lifetime Imaging Microscopy (FLIM) can distinguish between FITC signal and autofluorescence based on fluorescence decay rates.

  • Sequential scanning: For multi-labeled samples, use sequential rather than simultaneous scanning to reduce bleed-through.

  • Absorption analysis: Compare absorption spectra pre- and post-photobleaching; FITC signal will show characteristic changes while autofluorescence patterns differ.

  • Antibody validation approaches:

    • Gene knockdown/knockout controls

    • Peptide competition assays

    • Multiple antibodies to different epitopes

    • Correlation with protein expression by non-fluorescent methods

  • Image processing techniques:

    • Linear unmixing algorithms

    • Automated background subtraction using rolling ball algorithm

    • Tissue-specific autofluorescence reference libraries

  • Alternative detection strategy: Use non-fluorescent detection methods like chromogenic IHC in parallel to confirm localization patterns.

When presenting data from challenging samples, always include appropriate controls and raw unprocessed images alongside processed ones to allow independent evaluation of signal specificity.

How does site-specific FITC conjugation compare to traditional random conjugation for WWC3 antibody performance?

The method of conjugation can significantly impact antibody performance, particularly for targets requiring high precision in epitope recognition like WWC3:

Comparison of conjugation approaches:

AspectTraditional Random ConjugationSite-Specific Conjugation
ChemistryTargets lysine residues throughout antibody Directed to specific sites (e.g., engineered cysteines, glycans)
Conjugation UniformityHeterogeneous mixture of conjugatesHomogeneous preparation with defined conjugation sites
Impact on Antigen BindingPotential interference if conjugation occurs in Fab regionMinimal impact on binding when sites are away from CDRs
Batch-to-Batch ConsistencyVariableHighly reproducible
Optimization ComplexityRequires empirical optimization of reaction conditions Requires initial engineering but yields consistent results
Applicability to Various Antibody FormatsWorks with any antibody formatMay require antibody engineering or specific antibody formats

Methodological recommendations:

  • For research applications requiring ultimate sensitivity and consistency, site-specific conjugation offers advantages despite higher initial development costs.

  • For single-domain antibodies targeting WWC3, site-specific conjugation has demonstrated superior in vivo binding specificity compared to random conjugation methods, as shown in similar studies with other targets .

  • If using traditional random conjugation, optimize by:

    • Controlling buffer conditions (pH 9.0-9.5 typically optimal)

    • Using a range of FITC-to-antibody ratios in parallel reactions

    • Purifying and screening multiple conjugates for optimal performance

When selecting a conjugation approach, consider the specific research application, required sensitivity, and available resources.

What are the practical considerations for scaling up FITC conjugation of WWC3 antibodies for larger research studies?

Scaling up FITC conjugation reactions requires careful attention to several factors to maintain consistency and quality:

Key scaling considerations:

  • Antibody concentration consistency: Maintain the same antibody concentration (optimally ≥2 mg/ml) as the small-scale reactions to ensure comparable conjugation efficiency .

  • Buffer conditions: pH and buffer composition significantly impact conjugation efficiency. Use properly calibrated buffers and verify pH before and after scaling up.

  • Reaction vessel geometry: Surface-to-volume ratios affect reaction kinetics. Use vessels with similar geometry or adjust reaction times accordingly.

  • Mixing efficiency: Ensure adequate but gentle mixing throughout the reaction. Consider magnetic stirring for larger volumes rather than vortexing.

  • Temperature control: Maintain consistent temperature throughout the reaction, as temperature fluctuations can affect conjugation rates.

Practical workflow for scaled production:

  • Establish a reference standard from small-scale optimized reactions.

  • Implement quality control checkpoints at critical steps:

    • Pre-conjugation antibody quality (SEC-HPLC)

    • Post-conjugation DOL determination

    • Functional binding assays

  • Consider parallel processing of multiple smaller batches rather than a single large batch to manage risk.

  • Implement a staged purification approach for larger volumes:

    • Initial concentration step (if needed)

    • Buffer exchange using tangential flow filtration

    • Final polishing using appropriate chromatography

  • Validate each batch against the reference standard using:

    • Spectral properties

    • SDS-PAGE for conjugate homogeneity

    • Flow cytometry or ELISA for functional activity

This systematic approach helps maintain consistency when scaling up FITC conjugation processes for larger research studies.

What are the most common causes of batch-to-batch variability in FITC-conjugated antibodies and how can they be minimized?

Batch-to-batch variability can significantly impact experimental reproducibility. Understanding and addressing the causes of this variability is essential:

Common sources of variability and mitigation strategies:

Variability SourceImpactMitigation Strategy
Antibody quality variationDifferent proportions of active antibodyUse consistent antibody source; implement pre-conjugation quality checks
FITC reagent degradationReduced conjugation efficiencyUse fresh FITC vials; store desiccated at -20°C; prepare fresh solutions immediately before use
Inconsistent reaction conditionsVariable DOL and conjugation patternsStandardize temperature, time, pH, and mixing protocols; use calibrated equipment
Purification inconsistenciesVariable removal of free FITCStandardize column types, buffer compositions, and elution protocols
Storage condition differencesVariable degradation ratesImplement consistent aliquoting, storage containers, and temperatures

Standardization approach:

  • Implement a detailed batch record system documenting all parameters for each preparation.

  • Establish release criteria based on:

    • DOL (acceptable range)

    • Specific activity in standardized binding assay

    • Purity (% free FITC)

    • Spectral characteristics

  • Create reference standards from successful batches and use these for comparative analysis of new batches.

  • Consider implementing a Quality by Design (QbD) approach identifying Critical Quality Attributes (CQAs) and Critical Process Parameters (CPPs) that most impact final product quality.

By systematically controlling these variables, batch-to-batch consistency can be significantly improved, enhancing experimental reproducibility.

How should unexpected changes in FITC fluorescence intensity be investigated and resolved?

When encountering unexpected changes in FITC fluorescence intensity during experiments, a systematic troubleshooting approach is necessary:

Diagnostic flowchart for fluorescence intensity issues:

  • Verify instrument performance:

    • Run standardized fluorescent beads to check instrument calibration

    • Confirm laser alignment and power

    • Check filter sets for damage or incorrect installation

  • Assess sample preparation:

    • Review fixation protocol (overfixation can reduce signal)

    • Check permeabilization efficiency

    • Evaluate blocking effectiveness

  • Analyze antibody conjugate:

    • Check storage conditions and age of conjugate

    • Measure absorption spectrum to detect potential FITC degradation

    • Determine if precipitation has occurred (centrifuge and check pellet)

  • Examine buffer conditions:

    • Verify pH (FITC fluorescence decreases at acidic pH)

    • Check for presence of quenching agents

    • Test for presence of interfering substances (certain metal ions can quench fluorescence)

  • Review experimental protocol:

    • Antibody concentration appropriateness

    • Incubation times and temperatures

    • Washing stringency

Resolution strategies based on diagnosis:

  • For degraded conjugates: Prepare fresh conjugates or use newer aliquots

  • For pH issues: Adjust buffers to pH 7.5-8.5 for optimal FITC fluorescence

  • For quenching: Modify buffers to remove interfering components

  • For instrument issues: Recalibrate or service equipment

  • For protocol issues: Optimize critical parameters one at a time

Document all troubleshooting steps and outcomes to build an institutional knowledge base that can accelerate future troubleshooting efforts.

How are newer fluorophore technologies changing the landscape for applications traditionally using FITC-conjugated antibodies?

While FITC remains widely used, newer fluorophore technologies are expanding capabilities for researchers:

Emerging alternatives to FITC and their comparative advantages:

TechnologyAdvantages over FITCPotential Applications for WWC3 Studies
Quantum Dots20-30× brighter, extreme photostability, narrow emission spectraLong-term imaging, multiplexed detection
Fluorescent Proteins (genetic fusion)Direct expression in living systems, no conjugation requiredLive cell imaging of WWC3 interactions
Organic Dyes (Alexa/Atto/Dylight series)2-5× brighter than FITC, better photostability, pH insensitivityEnhanced sensitivity for low-abundance detection
Lanthanide ChelatesUltra-long Stokes shift, long fluorescence lifetimesTime-resolved imaging to eliminate autofluorescence
Click Chemistry-Compatible DyesSite-specific labeling, minimally modified antibodiesPrecise control over labeling location and DOL

Integration with advanced imaging techniques:

  • Super-resolution microscopy: Newer fluorophores compatible with STORM, PALM, and STED enable visualization of WWC3 localization at nanometer resolution, beyond the diffraction limit.

  • Multiplexed imaging: Spectral imaging systems combined with new fluorophores allow simultaneous detection of 10+ targets in the same sample.

  • Intravital imaging: Longer-wavelength fluorophores enable deeper tissue penetration for in vivo imaging applications.

  • Correlative microscopy: New dual-modality probes allow correlation between fluorescence microscopy and electron microscopy or mass spectrometry.

Researchers should evaluate these emerging technologies based on specific experimental requirements, including sensitivity needs, multiplexing requirements, and compatibility with existing systems.

What recent methodological advances have improved the sensitivity and specificity of antibody-based fluorescence detection that could be applied to WWC3 studies?

Recent methodological innovations have significantly enhanced the capabilities of fluorescence-based detection systems:

Advanced signal amplification strategies:

  • Proximity Ligation Assay (PLA): Detects protein-protein interactions with single-molecule sensitivity by generating fluorescent spots only when two proteins are in close proximity (<40 nm). Could reveal WWC3 interaction partners with unprecedented specificity.

  • Tyramide Signal Amplification (TSA): Enzymatic amplification that can increase sensitivity 10-100 fold compared to direct fluorophore conjugation. Particularly valuable for detecting low-abundance WWC3 in tissue sections.

  • DNA-PAINT: Uses transient binding of fluorophore-labeled DNA oligonucleotides for super-resolution imaging with exceptional signal-to-noise ratios. Could resolve WWC3 distribution at nanoscale precision.

Novel antibody formats and modifications:

  • Nanobodies/VHH fragments: Single-domain antibody fragments (~15 kDa vs. ~150 kDa for IgG) offer superior tissue penetration and resolution in microscopy. Evidence suggests improved performance in targeting specific epitopes .

  • Recombinant antibody fragments: Fab and scFv formats with site-specific fluorophore conjugation show improved orientation-controlled binding and reduced background.

  • Bispecific antibodies: Dual-targeting approaches can increase specificity by requiring recognition of two epitopes or combining target recognition with cell-type specificity.

Imaging and analysis innovations:

  • Artificial intelligence-based image analysis: Deep learning algorithms that can distinguish specific signals from background with greater accuracy than traditional thresholding approaches.

  • Spectral unmixing algorithms: Advanced computational methods that can separate fluorophore signals even with significant spectral overlap, enabling more precise multiplexed imaging.

  • Single-molecule detection techniques: Methods like coincidence detection that can identify true positive signals even at extremely low target abundance.

These methodological advances collectively offer significant improvements in sensitivity, specificity, and information content for WWC3 studies and other challenging research applications.

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.