APY6 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

The Anti-P2Y6 Receptor (extracellular)-FITC Antibody (#APR-106-F) is a fluorescein isothiocyanate (FITC)-conjugated reagent designed to detect the extracellular domain of the P2Y6 receptor. This receptor binds uridine diphosphate (UDP) and mediates intracellular calcium mobilization via Gq protein-coupled pathways .

Key Features:

  • Epitope: Targets the sequence (C)RNRTV(S)YDLSPPILS (residues 172–186) on the second extracellular loop of mouse P2RY6 .

  • Species Reactivity: Validated for human, rat, and mouse samples .

Table 1: Functional Roles of P2Y6

FunctionBiological ImpactSource
Calcium signalingMediates UDP-induced IP3/Ca²⁺ response
Vascular contractionFacilitates human cerebral artery contraction
Immune modulationExpressed in microglia and monocytes

Applications in Research

This antibody is widely used in multiple experimental workflows:

Table 2: Key Applications

ApplicationProtocol DetailsValidation Data
Western BlotDetects ~94 kDa band in lysates
ImmunohistochemistryLocalizes P2Y6 in fixed brain tissues
Live Cell Flow CytometryLabels surface P2Y6 on BV-2 microglia/THP-1 cells

4.1. Cell Surface Detection

  • In mouse BV-2 microglia, the antibody showed specific binding compared to isotype controls (2.5 µg dose) .

  • Similarly, in human THP-1 monocytes, it distinguished P2Y6-positive populations with minimal background .

4.2. Pathophysiological Relevance

  • P2Y6 activation mediates pro-inflammatory responses in microglia, linking it to neurodegenerative diseases .

  • Its role in cerebral artery contraction suggests involvement in vascular pathologies .

Validation and Quality Control

  • Specificity: Confirmed via knockout cell lines in Western blot and immunofluorescence .

  • Cross-reactivity: No reported cross-reactivity with other P2Y receptor subtypes .

Comparative Performance

The FITC-conjugated format (#APR-106-F) enables direct use in flow cytometry, eliminating secondary antibody requirements. Unconjugated versions (#APR-106) are optimal for multiplex assays .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 Weeks (Made-to-Order)
Synonyms
APY6 antibody; At2g02970 antibody; T17M13.14Probable apyrase 6 antibody; AtAPY6 antibody; EC 3.6.1.5 antibody; ATP-diphosphatase antibody; ATP-diphosphohydrolase antibody; Adenosine diphosphatase antibody; ADPase antibody; NTPDase antibody; Nucleoside triphosphate diphosphohydrolase 6 antibody
Target Names
APY6
Uniprot No.

Target Background

Function
This antibody targets an enzyme that catalyzes the hydrolysis of nucleoside tri- and diphosphate phosphoanhydride bonds. This enzyme is implicated in the regulation of pollen and anther development.
Database Links

KEGG: ath:AT2G02970

STRING: 3702.AT2G02970.1

UniGene: At.19550

Protein Families
GDA1/CD39 NTPase family
Subcellular Location
Cytoplasmic vesicle membrane; Multi-pass membrane protein.
Tissue Specificity
Detected in mature pollen grains (at the protein level). Also expressed in the veins and hydathode regions of rosette leaves.

Q&A

What is the APY6 antibody and how does it function in immunological research?

APY6 antibody, like other research antibodies, functions as an immunological tool for detecting and binding to specific target antigens. The antibody operates through the fundamental immunological principle where antibodies are proteins produced by B cells that bind to and inactivate antigens on pathogens or other targets of interest . In research applications, antibodies like APY6 can be used to visualize, identify, or isolate specific target molecules through their binding specificity. This occurs through the antibody's variable regions, particularly the complementarity-determining regions (CDRs), which determine the specific binding characteristics to the target epitope. The specificity of antibody-antigen interactions makes APY6 valuable in multiple research applications including immunohistochemistry, flow cytometry, western blotting, and immunoprecipitation techniques .

What validation methods should be used to confirm APY6 antibody specificity?

Validation of APY6 antibody specificity requires multiple complementary approaches to ensure reliable experimental results. Researchers should employ the following methodological sequence:

  • Positive and negative control testing: Use samples with known positive expression of the target antigen alongside samples where the target is known to be absent .

  • Cross-reactivity assessment: Test against similar antigens or closely related epitopes to determine if the antibody binds exclusively to its intended target .

  • Knockout/knockdown validation: Use genetic knockout or knockdown models where the target antigen is eliminated or reduced, which should correspondingly reduce or eliminate antibody binding .

  • Multiple antibody concordance: Compare results with other antibodies targeting the same antigen but recognizing different epitopes .

  • Orthogonal validation: Use non-antibody-based detection methods (e.g., mass spectrometry or PCR) to confirm expression levels of the target .

The validation process should be well-documented and included in research publications to ensure reproducibility of experimental findings across different research settings.

What are the optimal storage conditions to maintain APY6 antibody functionality?

Maintaining APY6 antibody functionality requires careful attention to storage conditions that preserve protein structure and prevent degradation. Optimal storage practices include:

  • Temperature maintenance: Store antibodies at -20°C for long-term storage or at 4°C for working solutions being used within 1-2 weeks .

  • Avoid freeze-thaw cycles: Repeated freezing and thawing can severely compromise antibody function through protein denaturation. Aliquot the antibody into single-use volumes before freezing .

  • Buffer considerations: Store in appropriate buffers containing stabilizers. Most commercial antibodies are formulated in phosphate-buffered saline (PBS) with preservatives such as sodium azide and stabilizing proteins like bovine serum albumin .

  • Light protection: For fluorophore-conjugated antibodies, protect from light exposure to prevent photobleaching .

  • Contamination prevention: Use sterile technique when handling antibodies to prevent microbial contamination, which can degrade the protein .

Researchers should maintain detailed records of storage conditions, freeze-thaw cycles, and lot numbers to track potential variability in experimental results.

How can computational approaches enhance APY6 antibody specificity prediction?

Advanced computational models can significantly improve APY6 antibody specificity prediction through several sophisticated approaches:

Biophysics-informed computational models can identify and disentangle multiple binding modes associated with specific ligands, allowing researchers to predict how APY6 antibodies might interact with various targets . These models operate by associating distinct binding modes with each potential ligand, enabling prediction beyond experimentally observed interactions .

The implementation process involves:

  • Data collection: Training the model on a set of experimentally selected antibodies with known binding characteristics .

  • Binding mode identification: Computational analysis to identify distinct binding modes associated with different ligands .

  • Energy function optimization: For specific binding profiles, jointly minimizing the energy functions associated with desired ligands while maximizing those associated with undesired ligands .

  • Sequence optimization: Generating novel antibody sequences with predefined binding profiles by optimizing over energy functions .

This computational approach has demonstrated success in designing antibodies with both specific and cross-specific binding properties, allowing researchers to develop APY6 antibody variants with customized specificity profiles even for chemically similar ligands that cannot be experimentally dissociated .

What methodological approaches can resolve cross-reactivity issues with APY6 antibody?

Resolving cross-reactivity issues with APY6 antibody requires a comprehensive methodological approach combining experimental and computational techniques:

  • Epitope mapping: Precisely identify the binding epitope using techniques such as hydrogen-deuterium exchange mass spectrometry (HDX-MS), X-ray crystallography, or cryo-electron microscopy to understand the molecular basis of cross-reactivity .

  • Competitive binding assays: Perform assays with graduated concentrations of potential cross-reactive antigens to quantify the relative binding affinities and identify problematic epitopes .

  • Sequence analysis and mutagenesis: Analyze the antibody variable region sequences to identify critical residues involved in binding, followed by site-directed mutagenesis to improve specificity .

  • Computational redesign: Apply biophysics-informed models that can minimize binding energy for desired targets while maximizing it for undesired cross-reactive targets . This approach has been successfully demonstrated in engineering antibodies with customized specificity profiles .

  • Machine learning integration: Implement machine learning algorithms trained on existing antibody-antigen interaction data to predict modifications that might enhance specificity .

  • Phage display refinement: Use phage display with negative selection strategies against cross-reactive antigens to isolate variants with improved specificity profiles .

The combination of these approaches has shown success in generating antibodies with both specific and cross-specific binding properties while mitigating experimental artifacts and biases in selection experiments .

How can artificial intelligence technologies improve the development of APY6 antibody variants?

Artificial intelligence technologies are revolutionizing antibody development through several advanced methodologies applicable to APY6 antibody research:

Vanderbilt University Medical Center's approach, supported by up to $30 million from ARPA-H, demonstrates the potential of AI in antibody development by building a massive antibody-antigen atlas and developing AI-based algorithms to engineer antigen-specific antibodies . This method addresses traditional bottlenecks in antibody discovery, including inefficiency, high costs, logistical hurdles, and limited scalability .

The AI-enhanced development process for APY6 antibody variants involves:

  • Data compilation: Building comprehensive antibody-antigen interaction databases that serve as training datasets for AI models .

  • Binding mode identification: Using machine learning to identify distinct binding modes associated with different antigens .

  • Sequence-structure-function modeling: Implementing deep learning models that can predict how changes in antibody sequence affect structure and binding function .

  • In silico optimization: Using computational methods to optimize antibody sequences for specific binding profiles before experimental validation .

  • Democratized discovery process: Creating accessible platforms that allow researchers to generate monoclonal antibody therapeutics against specific targets more efficiently .

This AI-driven approach enables the generation of antibody variants with customized binding profiles, either with high specificity for particular targets or with cross-specificity for multiple target ligands, expanding the potential research applications of APY6 antibody technology .

What standardized protocols should be used for APY6 antibody in different immunoassay applications?

Standardized protocols for APY6 antibody applications must be carefully optimized for each specific immunoassay technique to ensure reproducible and reliable results:

For Western Blotting:

  • Sample preparation: Optimize protein extraction buffers to preserve the target antigen's native conformation.

  • Blocking optimization: Test multiple blocking agents (BSA, non-fat milk, commercially available blockers) at various concentrations to determine optimal signal-to-noise ratio.

  • Antibody concentration: Perform titration experiments (typically starting at 1:1000 dilution) to determine optimal antibody concentration that maximizes specific signal while minimizing background.

  • Incubation conditions: Standardize incubation time and temperature (typically 1-2 hours at room temperature or overnight at 4°C).

  • Washing stringency: Adjust washing buffer composition (TBST or PBST) and number of washes to remove non-specific binding .

For Immunohistochemistry/Immunofluorescence:

  • Fixation method: Compare paraformaldehyde, methanol, and acetone fixation to determine which best preserves the target epitope.

  • Antigen retrieval: Test multiple retrieval methods (heat-induced epitope retrieval at various pH levels, enzymatic retrieval) to optimize epitope accessibility.

  • Permeabilization: Adjust detergent concentration (typically 0.1-0.3% Triton X-100) based on target subcellular localization.

  • Controls: Include isotype controls and target-negative tissues to validate staining specificity .

Each application requires detailed documentation of optimization steps to ensure reproducibility across different research settings.

How should researchers interpret conflicting APY6 antibody experimental results?

When faced with conflicting experimental results using APY6 antibody, researchers should implement a systematic troubleshooting approach:

  • Analytical validation assessment: Re-examine the validation data for the antibody lot being used. Different lots may have varying specificity and sensitivity characteristics .

  • Experimental condition comparison: Create a detailed comparison table documenting all experimental variables between conflicting experiments, including:

    VariableExperiment 1Experiment 2Potential Impact
    Antibody lot#12345#12346Lot-to-lot variability
    Antibody concentration1:10001:500Signal intensity differences
    Incubation timeOvernight 4°C2h RTBinding equilibrium
    Sample preparationRIPA bufferNP-40 bufferEpitope accessibility
    Detection methodChemiluminescenceFluorescenceSensitivity differences
  • Independent confirmation: Use orthogonal methods that don't rely on antibody binding (e.g., mass spectrometry, PCR) to verify target expression levels .

  • Biological variation consideration: Evaluate whether conflicting results reflect actual biological variability rather than technical issues .

  • Epitope accessibility analysis: Determine if sample preparation methods differently affect epitope conformation or accessibility .

  • Cross-reactivity assessment: Investigate potential cross-reactivity with structurally similar proteins in the experimental system .

By systematically addressing these factors and documenting the investigation process, researchers can identify the source of conflicting results and establish reliable experimental conditions for future studies.

What database resources can be used to compare APY6 antibody sequences with other characterized antibodies?

Researchers can leverage several specialized database resources to compare APY6 antibody sequences with other characterized antibodies:

The Observed Antibody Space (OAS) database provides a comprehensive resource of cleaned and annotated antibody sequences that can be used for comparative analysis . This database now contains 1.5 billion unpaired sequences from 80 studies and paired sequencing data from five studies, allowing researchers to search for sequences with the same V and J genes as the APY6 antibody .

Key database resources and their applications include:

  • Observed Antibody Space (OAS): Provides standardized, annotated antibody sequences with both nucleotides and amino acids, allowing for sequence-based searches and comparison. The web interface (http://opig.stats.ox.ac.uk/webapps/oas/) enables researchers to quickly identify antibodies with similar structural features to APY6 .

  • ImmuneAccess: Contains a large set of annotated CDR3 sequences that can be used to compare APY6's CDR3 region with other antibodies .

  • RAPID (Rep-seq dataset analysis platform): Offers identically processed human antibody sequences for standardized comparison .

  • AIRR Data Commons (ADC): Provides access to a network of geographically distributed AIRR-compliant repositories through a single API, expanding the comparison dataset .

  • PIRD (Pan Immune Repertoire Database): Contains large collections of annotated BCR-seq data that can provide insight into the frequency and distribution of sequences similar to APY6 .

Using these resources, researchers can:

  • Identify antibodies with similar binding characteristics

  • Analyze sequence conservation in key binding regions

  • Evaluate the prevalence of similar sequences in different populations

  • Guide engineering efforts to improve APY6 specificity or affinity

What are the most efficient methods for determining APY6 antibody binding kinetics?

Determining binding kinetics for APY6 antibody requires precise methodologies that can quantify association and dissociation rates. The most efficient methods include:

  • Surface Plasmon Resonance (SPR): This label-free technique allows real-time measurement of binding kinetics by immobilizing either the antibody or antigen on a sensor chip and flowing the binding partner over the surface . Key parameters to optimize include:

    • Surface density of the immobilized component

    • Flow rate to minimize mass transport limitations

    • Buffer composition to reduce non-specific binding

    • Multiple analyte concentrations for accurate model fitting

  • Bio-Layer Interferometry (BLI): Similar to SPR but using optical interference patterns to measure binding, BLI offers advantages in terms of throughput and sample consumption . Important considerations include:

    • Sensor selection based on immobilization chemistry

    • Sample concentration range spanning the expected KD

    • Optimization of association and dissociation times

  • Isothermal Titration Calorimetry (ITC): Measures the heat released or absorbed during binding to determine thermodynamic parameters alongside kinetics . This provides valuable information about the enthalpy and entropy contributions to binding.

  • Microscale Thermophoresis (MST): Detects changes in the movement of molecules along temperature gradients upon binding, requiring minimal sample amounts and offering high sensitivity .

These techniques collectively provide a comprehensive profile of APY6 binding characteristics, with typical data reporting:

  • Association rate constant (ka)

  • Dissociation rate constant (kd)

  • Equilibrium dissociation constant (KD)

  • Thermodynamic parameters (ΔH, ΔS, ΔG)

This information is crucial for comparing APY6 with other antibodies and predicting its performance in various research applications.

How can epitope mapping be optimized for APY6 antibody target recognition studies?

Optimizing epitope mapping for APY6 antibody requires a multi-technique approach to comprehensively characterize its target recognition properties:

  • X-ray Crystallography and Cryo-EM: These structural biology approaches provide atomic-level resolution of the antibody-antigen complex, revealing precise binding interfaces . While resource-intensive, they offer unparalleled detail about the structural basis of recognition.

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): This approach identifies regions of the antigen protected from deuterium exchange when bound by the antibody, indicating binding sites. Optimization requires:

    • Careful control of exchange times

    • Optimization of proteolytic digestion conditions

    • Comprehensive peptide mapping of the antigen

    • Comparison of exchange rates between bound and unbound states

  • Alanine Scanning Mutagenesis: Systematically replacing amino acids in the suspected epitope region with alanine to identify critical binding residues. This approach can be enhanced by:

    • Using computational predictions to prioritize residues for mutation

    • Creating a comprehensive substitution matrix

    • Quantifying binding changes with SPR or ELISA

  • Peptide Arrays or Phage Display Libraries: These high-throughput approaches can map linear epitopes by testing antibody binding to overlapping peptides or peptide libraries displayed on phages .

  • Computational Epitope Prediction: Bioinformatics approaches can predict epitopes based on structural features and sequence analysis, guiding experimental mapping efforts .

Integration of multiple mapping techniques is essential for accurate characterization, as each method has inherent limitations and biases. Triangulation of results across methods provides the most reliable epitope identification.

What quality control metrics should be established for APY6 antibody batch-to-batch consistency?

Establishing rigorous quality control metrics for APY6 antibody ensures experimental reproducibility and reliability across different production batches:

A comprehensive quality control protocol should include the following critical metrics:

  • Binding Affinity Determination: Each batch should demonstrate consistent KD values (within ±20% of reference batch) as measured by SPR or ELISA. Test against multiple concentrations of target antigen to generate complete binding curves .

  • Specificity Profile Assessment: Cross-reactivity testing against structurally similar antigens should show consistent patterns across batches. Implement:

    • Side-by-side testing with reference batch

    • Competitive binding assays

    • Testing against a panel of potential cross-reactants

  • Physical Characterization:

    • Size Exclusion Chromatography (SEC) to monitor aggregation (<5% acceptable)

    • Isoelectric focusing to verify charge profile consistency

    • SDS-PAGE for purity assessment (>95% purity required)

    • Mass spectrometry for intact mass verification and detection of post-translational modifications

  • Functional Performance in Applications:

    • Standardized western blot with reference samples

    • Immunohistochemistry on validated positive and negative tissues

    • Flow cytometry with target-expressing and non-expressing cell lines

  • Stability Assessment:

    • Accelerated stability testing (e.g., 1 week at 37°C)

    • Real-time stability monitoring

    • Freeze-thaw cycle tolerance testing

Documentation should include detailed batch records with acceptance criteria for each parameter. Implementation of a reference standard (master batch) against which all new batches are compared ensures long-term consistency and enables detection of manufacturing drift over time.

How can APY6 antibody be effectively used in immunoprecipitation-mass spectrometry studies?

Effectively employing APY6 antibody in immunoprecipitation-mass spectrometry (IP-MS) studies requires careful optimization of multiple experimental parameters:

  • Antibody Immobilization Strategy: Compare multiple coupling approaches:

    • Direct covalent conjugation to beads (using NHS-ester chemistry)

    • Indirect capture via Protein A/G beads

    • Biotinylated antibody with streptavidin support

    Each method may differentially preserve antibody orientation and binding capacity. Quantify recovery of known targets to determine optimal approach .

  • Lysis and Buffer Optimization: Test multiple lysis conditions to maximize target solubilization while maintaining antibody-target interaction:

    • RIPA buffer (strong solubilization but potentially disruptive)

    • NP-40 buffer (milder, better for preserving protein complexes)

    • Digitonin (gentlest, best for protein complexes)

    Include protease and phosphatase inhibitors to preserve target integrity .

  • Pre-clearing Strategy: Implement stringent pre-clearing using control IgG of the same species to reduce non-specific binding:

    • Use matched concentration of control antibody

    • Perform pre-clearing at the same temperature as IP

    • Match incubation time to actual IP

  • Washing Optimization: Develop a graduated washing strategy with increasing stringency:

    • Initial washes with IP buffer to remove loosely bound contaminants

    • Intermediate washes with slightly increased salt concentration

    • Final washes with MS-compatible buffers to remove detergents

  • Elution Conditions: Compare multiple elution strategies for compatibility with downstream MS analysis:

    • Gentle: Competitive elution with excess antigen peptide

    • Moderate: Low pH glycine buffer

    • Stronger: SDS or urea-based buffers (require additional cleanup)

  • MS Sample Preparation: Optimize digestion and peptide recovery:

    • In-solution vs. in-gel digestion comparison

    • Filter-aided sample preparation (FASP) for detergent removal

    • Stage tip purification for salt removal

This methodical approach maximizes the specificity of APY6 antibody in IP-MS applications while minimizing background and ensuring compatibility with sensitive MS detection.

What considerations are important when using APY6 antibody for multiplexed imaging techniques?

Implementing APY6 antibody in multiplexed imaging techniques requires careful consideration of several technical parameters:

  • Antibody Labeling Optimization:

    • Compare different fluorophores or detection tags to identify those with minimal impact on binding affinity

    • Determine optimal fluorophore-to-antibody ratio (FAR) by testing multiple conjugation conditions

    • Confirm retained specificity after labeling through side-by-side comparison with unconjugated antibody

  • Panel Design for Multiplex Studies:

    • Spectral compatibility assessment to minimize bleed-through between channels

    • Strategic epitope selection when combining multiple antibodies to avoid steric hindrance

    • Hierarchical staining sequence optimization based on antibody sensitivity to fixation and retrieval conditions

  • Cross-Reactivity Mitigation:

    • Thorough cross-reactivity testing against all antigens in the multiplexed panel

    • Implementation of blocking steps between sequential staining rounds

    • Validation of staining patterns in single-stained controls versus multiplexed samples

  • Signal Amplification Considerations:

    • Tyramide signal amplification (TSA) optimization if APY6 targets low-abundance antigens

    • Careful titration of detection reagents to prevent signal saturation or spillover

    • Use of appropriate controls to distinguish specific signal from autofluorescence

  • Cyclic Immunofluorescence Implementation:

    • Antibody elution condition optimization to ensure complete removal between cycles

    • Validation of epitope preservation across multiple staining/elution cycles

    • Image registration strategies for accurate overlay of sequential staining rounds

  • Image Analysis Workflow Development:

    • Cell segmentation algorithm optimization for tissue context

    • Background subtraction methods appropriate for tissue autofluorescence

    • Quantification strategy for co-localization analysis

These considerations ensure APY6 antibody performs optimally in complex multiplexed imaging settings while maintaining specificity and sensitivity across different experimental conditions.

What strategies can resolve non-specific binding issues with APY6 antibody?

Resolving non-specific binding issues with APY6 antibody requires a systematic approach addressing multiple potential causes:

  • Blocking Optimization:

    • Compare multiple blocking agents (BSA, casein, normal serum, commercial blockers) at various concentrations

    • Extend blocking time (1-2 hours at room temperature to overnight at 4°C)

    • Test species-matched normal serum for blocking when using secondary detection systems

  • Buffer Modification Strategy:

    • Increase detergent concentration incrementally (0.1% to 0.3% Tween-20 or Triton X-100)

    • Add carrier proteins (0.1-1% BSA or gelatin) to reduce non-specific interactions

    • Adjust salt concentration (150mM to 500mM NaCl) to disrupt weak ionic interactions

    • Test additives like 5% glycerol or 1-5mM EDTA to reduce non-specific binding

  • Antibody Concentration Titration:

    • Perform systematic dilution series to identify optimal concentration

    • Compare signal-to-noise ratio across concentration range

    • Consider prolonged incubation with more dilute antibody rather than shorter incubation with concentrated antibody

  • Pre-adsorption Protocol Implementation:

    • Pre-incubate diluted antibody with tissues or cells lacking the target

    • Use control recombinant proteins to compete away non-specific binding

    • Implement immunodepletion against known cross-reactive antigens

  • Secondary Detection System Modification:

    • Test F(ab')2 fragments instead of whole IgG to reduce Fc-mediated binding

    • Compare different detection systems (direct labeling vs. biotin-streptavidin vs. polymer detection)

    • Use species-specific secondary antibodies with minimal cross-reactivity

  • Sample Preparation Refinement:

    • Optimize fixation conditions to better preserve epitope structure

    • Test alternative antigen retrieval methods

    • Implement additional quenching steps for endogenous enzyme activity or fluorescence

Systematic testing of these parameters through controlled experiments allows identification of the specific factors contributing to background, enabling development of an optimized protocol with maximum signal-to-noise ratio.

How can researchers validate APY6 antibody for use in emerging technologies like spatial transcriptomics?

Validating APY6 antibody for emerging technologies like spatial transcriptomics requires specialized approaches that integrate protein detection with transcriptomic analysis:

  • Orthogonal Validation Strategy:

    • Correlate APY6 antibody staining patterns with mRNA expression of the target gene using methods like RNAscope or in situ hybridization

    • Verify concordance between protein and mRNA localization patterns

    • Perform quantitative analysis of spatial correlation between transcripts and protein signals

  • Compatibility Assessment:

    • Test antibody performance under fixation and permeabilization conditions required for nucleic acid retention

    • Validate staining after exposure to enzymatic treatments used in spatial transcriptomics workflows

    • Ensure signal persistence through multiple imaging cycles if sequential detection is required

  • Multiplexing Validation:

    • Confirm antibody performance in presence of oligonucleotide-conjugated detection reagents

    • Test for potential interference with hybridization steps or nucleic acid amplification

    • Optimize signal amplification to match sensitivity of transcriptomic detection

  • Cross-Platform Comparison:

    • Implement side-by-side validation across complementary spatial biology platforms

    • Compare results between antibody-based and transcript-based detection of the same target

    • Quantify concordance rates and investigate discrepancies

  • Computational Validation Framework:

    • Develop analysis pipelines that integrate protein and transcript data

    • Implement statistical methods to assess correlation between modalities

    • Apply machine learning approaches to identify patterns of protein-mRNA relationships

  • Reference Standards Implementation:

    • Use control samples with known expression patterns as validation benchmarks

    • Include spike-in controls with predetermined ratios of protein and mRNA

    • Employ tissue microarrays with graduated expression levels for quantitative validation

This comprehensive validation approach ensures that APY6 antibody can be reliably integrated into multimodal spatial biology workflows, providing confident protein detection that complements spatial transcriptomic data.

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.