CAT3 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CAT3 Antibody

The CAT3 antibody specifically targets the cationic amino acid transporter 3 (CAT3), encoded by the SLC7A3 gene. CAT3 is a member of the solute carrier family 7 (SLC7), which facilitates the transport of cationic amino acids such as arginine, lysine, and ornithine across cell membranes. This antibody is primarily used in research to study CAT3's expression, localization, and functional roles in physiological and pathological processes, including hematopoietic stem cell (HSC) regulation and immune responses .

Antibody Structure

  • Target antigen: Human SLC7A3 (UniProt ID: Q8WY07), a 67 kDa transmembrane protein .

  • Host species: Rabbit-derived polyclonal antibody .

  • Epitope: Recognizes the C-terminal region of SLC7A3 .

Biological Role of CAT3

  • Function: Facilitates arginine uptake, critical for cellular metabolism and immune cell activation .

  • Tissue distribution: Predominantly expressed in the thymus, brain, and hematopoietic tissues .

Key Validation Data

ParameterDetails
Western BlotDetects a band at ~70 kDa in human MOLT-4 and Daudi cell lysates .
SpecificityValidated using SLC7A3 knockout models; no cross-reactivity observed .
Positive ControlsHuman MOLT-4 (T-cell leukemia) and Daudi (Burkitt’s lymphoma) cells .

Research Applications

  1. Protein Expression Analysis: Used to quantify CAT3 levels in immune and cancer cell lines .

  2. Functional Studies: Investigates CAT3's role in arginine-dependent processes, such as HSC maintenance and T-cell activation .

  3. Disease Models: Applied in studies of leukemogenesis and metabolic disorders .

CAT3 in Hematopoiesis

  • Knockout Studies: Slc7a3 knockout mice showed normal hematopoiesis and T-cell activation, suggesting compensatory mechanisms by other transporters (e.g., CAT1/CAT2) .

  • Metabolic Role: Extracellular arginine is critical for HSC function, but CAT3 is dispensable under homeostatic conditions .

Technical Insights

  • Antibody Performance: Exhibits high affinity in Western blot (0.25–0.5 µg/mL working concentration) and compatibility with reducing SDS-PAGE conditions .

Comparative Analysis of CAT3 Antibody Clones

Clone/ProductHostApplicationsKey Features
A09720-3 (Boster)RabbitWB, IHC, ELISAValidated in leukemia/lymphoma models .
Commercial AlternativesMouseFlow cytometryTargets extracellular CAT3 domains (not listed in provided sources).

Limitations and Future Directions

  • Limitations: Limited data on CAT3’s role in cancer progression or immune evasion.

  • Opportunities: Development of monoclonal CAT3 antibodies for therapeutic targeting (e.g., arginine-depletion strategies in tumors) .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
CAT3 antibody; At1g20620 antibody; F2D10.40 antibody; F5M15.5Catalase-3 antibody; EC 1.11.1.6 antibody
Target Names
CAT3
Uniprot No.

Target Background

Function

Catalase 3 (CAT3) is a ubiquitous enzyme in aerobically respiring organisms. Its primary function is to protect cells from the damaging effects of hydrogen peroxide.

Gene References Into Functions

Relevant studies have elucidated several key aspects of CAT3 function and regulation:

  1. The host proteasome pathway plays a role in the degradation of CAT3, particularly within tissues infected by cytomegalovirus (CMV). PMID: 27659495
  2. Direct interaction between CMV 2b protein and CAT3 has been observed in infected tissues. PMID: 21622812
  3. CAT3 expression increases with age, correlating with elevated hydrogen peroxide levels in vascular bundles. PMID: 17080932
Database Links

KEGG: ath:AT1G20620

STRING: 3702.AT1G20620.1

UniGene: At.24821

Protein Families
Catalase family
Subcellular Location
Peroxisome.

Q&A

What is CAT3/SLC7A3 and why is it targeted in immunological research?

CAT3 (Cationic Amino Acid Transporter 3), also known as SLC7A3 (Solute Carrier Family 7 Member 3), is a membrane protein that functions as a uniporter mediating the uptake of cationic L-amino acids including L-arginine, L-lysine, and L-ornithine. In humans, the canonical protein consists of 619 amino acid residues with a molecular weight of approximately 67.2 kDa . It is primarily localized to the cell membrane and demonstrates high expression in specific tissues including the thymus, uterus, and testis .

CAT3 is targeted in immunological research due to its role in regulating amino acid transport, which can impact various cellular functions including protein synthesis, nitric oxide production, and immune response mechanisms. Additionally, its interaction with viral proteins, such as the Cucumber mosaic virus (CMV) 2b protein, has been documented to affect host defense mechanisms, making it relevant for studies on host-pathogen interactions .

What are the most effective validation methods for CAT3 antibodies before experimental use?

When validating CAT3 antibodies for experimental use, researchers should implement a multi-step approach:

  • Western Blot Validation: Confirm specific binding to the target protein at the expected molecular weight (67.2 kDa for human CAT3) . Include appropriate positive controls (tissues known to express CAT3 such as thymus, uterus, or testis) and negative controls (tissues with minimal CAT3 expression or CAT3 knockout samples if available).

  • Knockout/Knockdown Validation: Compare antibody signal between wild-type samples and CAT3 knockout or knockdown samples. A significant reduction in signal in the knockout/knockdown samples confirms specificity .

  • Peptide Competition Assay: Pre-incubate the antibody with a synthetic peptide corresponding to the immunogen. If the antibody is specific, this should substantially reduce or eliminate the signal in subsequent applications.

  • Cross-reactivity Testing: Evaluate potential cross-reactivity with other CAT family members (CAT1/SLC7A1, CAT2/SLC7A2) due to sequence homology.

  • Immunoprecipitation Followed by Mass Spectrometry: For advanced validation, perform IP with the antibody followed by mass spectrometry to confirm that the precipitated protein is indeed CAT3.

What are the optimal sample preparation protocols for detecting CAT3 in different tissue types?

The optimal sample preparation protocols for detecting CAT3 vary by tissue type and application:

For Thymus, Uterus, and Testis (High-expression Tissues) :

  • Harvest fresh tissue and immediately place in ice-cold PBS.

  • For protein extraction, homogenize in RIPA buffer supplemented with protease inhibitors.

  • Include detergents like 0.1% SDS and 1% Triton X-100 to effectively solubilize membrane-bound CAT3.

  • Centrifuge at 14,000 × g for 15 minutes at 4°C to remove cell debris.

  • Quantify protein concentration using BCA or Bradford assay.

  • Store aliquots at -80°C to avoid freeze-thaw cycles.

For Tissues with Lower CAT3 Expression:

  • Consider using membrane fractionation techniques to enrich for membrane proteins.

  • Use mild detergents (0.5% NP-40 or 1% digitonin) for initial extraction to maintain protein conformation.

  • For detection by immunohistochemistry, optimize antigen retrieval conditions (typically citrate buffer pH 6.0 or EDTA buffer pH 9.0).

For Cell Line Models:

  • Culture cells to 80-90% confluence before harvesting.

  • Include phosphatase inhibitors if analyzing phosphorylation status.

  • Consider crosslinking protocols if studying protein-protein interactions.

Important Considerations:

  • As CAT3 undergoes N-glycosylation , enzymatic deglycosylation may be necessary for certain applications to reduce heterogeneity in protein migration patterns.

  • Proteasome inhibitors should be included if studying CAT3 degradation, as evidence suggests involvement of the ubiquitin-proteasome pathway in CAT3 regulation .

How do post-translational modifications of CAT3 affect antibody recognition, and how can researchers account for this?

Post-translational modifications (PTMs) of CAT3, particularly N-glycosylation , can significantly impact antibody recognition in multiple ways:

Effects on Antibody Recognition:

  • Epitope Masking: Glycosylation can physically block antibody access to protein epitopes, particularly if the epitope is adjacent to or contains glycosylation sites.

  • Altered Protein Conformation: PTMs may induce conformational changes that affect exposure of conformational epitopes.

  • Migration Pattern Variability: Glycosylated CAT3 shows heterogeneous migration patterns in gel electrophoresis, potentially complicating band interpretation.

  • Tissue-Specific Modification Patterns: The glycosylation pattern of CAT3 may vary across tissues, leading to differential antibody reactivity.

Strategies to Account for PTM Effects:

StrategyMethodologyApplicationsLimitations
DeglycosylationTreat samples with PNGase F or Endoglycosidase H before analysisWestern blot, IPMay denature proteins; not suitable for live-cell studies
PTM-specific antibodiesUse antibodies specifically targeting glycosylated or non-glycosylated formsDetecting specific CAT3 variantsLimited commercial availability
Multiple antibody approachUse antibodies targeting different epitopesCross-validation of resultsIncreases experimental complexity and cost
Mass spectrometryCharacterize PTM patterns before antibody selectionDetailed PTM mappingResource-intensive; requires specialized equipment

Practical Recommendations:

  • When studying CAT3 in contexts where PTM status is critical, perform parallel experiments with and without deglycosylation treatment.

  • Document the exact band pattern observed in your experimental system to establish a reliable reference.

  • For interaction studies, consider whether the interaction depends on PTM status, particularly since the ubiquitin-proteasome pathway has been implicated in CAT3 regulation .

What are the most effective strategies for studying CAT3 interaction with viral proteins, particularly the CMV 2b protein?

Studying the interaction between CAT3 and viral proteins such as the CMV 2b protein requires specialized approaches due to the complex nature of these interactions and their biological significance :

Recommended Strategies:

  • Co-immunoprecipitation (Co-IP) Optimization:

    • Use anti-CAT3 antibodies for pull-down followed by detection of viral proteins, or vice versa.

    • Include appropriate detergent conditions (typically 0.5-1% NP-40) to maintain membrane protein solubility without disrupting interactions.

    • Consider crosslinking with DSP or formaldehyde to stabilize transient interactions.

    • Always include negative controls using non-specific antibodies of the same isotype.

  • Proximity Ligation Assay (PLA):

    • Particularly valuable for detecting CAT3-viral protein interactions in situ.

    • Requires specific antibodies raised in different species.

    • Provides spatial information about the interaction in cellular contexts.

  • FRET/BRET Analysis:

    • Generate fluorescent or bioluminescent protein fusions of CAT3 and viral proteins.

    • Allows real-time monitoring of interactions in live cells.

    • Important to validate that fusion proteins retain native localization and function.

  • BiFC (Bimolecular Fluorescence Complementation):

    • Fuse complementary fragments of fluorescent proteins to CAT3 and viral proteins.

    • Reconstitution of fluorescence indicates proximity/interaction.

    • Useful for mapping interaction domains when combined with truncation mutants.

  • Functional Impact Assessment:

    • Compare viral replication in wild-type versus CAT3 knockout/knockdown systems .

    • Measure H₂O₂ levels and oxidative stress markers to assess functional consequences of the interaction.

    • Analyze necrosis phenotypes in plant models as functional readouts of the interaction .

Critical Considerations:

  • The interaction between CAT3 and viral proteins may be indirect or part of larger complexes; therefore, confirmatory approaches using multiple techniques are recommended.

  • The ubiquitin-proteasome pathway involvement suggests that proteasome inhibitors should be included when studying the stability aspects of this interaction .

  • When using antibodies in these interaction studies, epitope accessibility in the context of the CAT3-viral protein complex should be assessed, as the interaction may mask antibody binding sites.

How can researchers distinguish between specific and non-specific binding when using CAT3 antibodies in complex tissue samples?

Distinguishing between specific and non-specific binding is crucial for accurate interpretation of CAT3 antibody results, especially in complex tissue samples:

Advanced Validation Approaches:

  • Orthogonal Antibody Validation:

    • Use multiple antibodies targeting different epitopes of CAT3.

    • Concordant results across different antibodies strongly indicate specific binding.

    • Create a validation matrix documenting the performance of each antibody across applications.

  • Genetic Controls:

    • Compare staining patterns between wild-type and CAT3 knockout/knockdown samples .

    • Utilize CRISPR-Cas9 edited cell lines with tagged endogenous CAT3 as positive controls.

    • Employ siRNA-mediated knockdown with dose-dependent reduction in signal as evidence of specificity.

  • Advanced Imaging Techniques:

    • Super-resolution microscopy to confirm expected subcellular localization (cell membrane) .

    • Colocalization studies with known membrane markers.

    • FRAP (Fluorescence Recovery After Photobleaching) to assess dynamics consistent with a membrane transporter.

  • Biochemical Validations:

    • Peptide competition assays with titrated amounts of blocking peptide.

    • Immunodepletion experiments to confirm antibody exhaustion correlates with signal reduction.

    • Mass spectrometry analysis of immunoprecipitated material to confirm target identity.

Quantitative Assessment Methods:

MethodMetricsThreshold for Specificity
Signal-to-Noise RatioMean specific signal / Mean background signal>3:1 for reasonable specificity; >10:1 for high confidence
Knockdown Efficiency CorrelationCorrelation coefficient between knockdown level and signal reductionr > 0.8 indicates high specificity
Z-score(Sample signal - Negative control signal) / Standard deviation of negative controlZ > 3 suggests specific binding
Mander's Overlap CoefficientFor colocalization with known CAT3 markers>0.7 indicates significant overlap

Protocol Adjustments to Minimize Non-specific Binding:

  • Optimize blocking conditions using 5% BSA or 5% non-fat dry milk in TBS-T.

  • Include competitive blocking agents such as normal serum from the same species as the secondary antibody.

  • Perform stringent washing steps (3-5 washes of 5-10 minutes each).

  • For IHC/ICC, include an avidin/biotin blocking step if using biotinylated secondary antibodies.

  • Pre-adsorb antibodies against tissues from CAT3 knockout models when available.

What methodological approaches are most effective for studying CAT3's role in viral infection mechanisms?

Investigating CAT3's role in viral infection mechanisms requires integrated approaches that combine molecular, cellular, and physiological techniques:

Recommended Methodological Framework:

  • Infection Model Establishment:

    • Compare viral replication kinetics in wild-type versus CAT3-knockout systems .

    • Develop cell line models with varying CAT3 expression levels to establish dose-dependency.

    • Consider using primary cells from tissues with high CAT3 expression (thymus, uterus, testis) for physiological relevance.

  • Interaction Characterization:

    • Implement time-course co-immunoprecipitation studies to track CAT3-viral protein complex formation during infection progression.

    • Use FRET/FLIM approaches for live-cell monitoring of interactions.

    • Apply hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces at molecular resolution.

  • Functional Readouts:

    • Measure amino acid transport activity using radiolabeled substrates (L-arginine, L-lysine, L-ornithine) in infected versus uninfected cells.

    • Quantify H₂O₂ levels and oxidative stress markers as indicators of functional consequences .

    • Assess necrosis development as a phenotypic readout in appropriate model systems .

  • Mechanistic Dissection:

    • Analyze ubiquitination patterns of CAT3 during infection using ubiquitin-specific antibodies and proteasome inhibitors .

    • Perform ribosome profiling to assess impacts on global and virus-specific translation.

    • Investigate nitric oxide production changes, given CAT3's role in transporting arginine (a precursor for NO synthesis) .

Advanced Experimental Design Considerations:

Experimental ApproachKey MeasurementsTechnical Considerations
Time-course proteomicsCAT3 levels and PTM changes during infectionInclude proteasome inhibitors; focus on membrane fractions
Proximity-dependent biotin identification (BioID)CAT3 interactome changes upon infectionUse viral protein-BioID fusions; compare to uninfected controls
CRISPR screensViral replication dependency factors in CAT3 pathwaysDesign sgRNA libraries targeting CAT3-interacting proteins
Targeted metabolomicsAmino acid pools and fluxFocus on CAT3 substrates (arginine, lysine, ornithine)
Single-cell RNA-seqHeterogeneity in response to infectionCompare CAT3-high vs. CAT3-low cells within population

Integration with Antibody Applications:

  • Use phospho-specific antibodies to track CAT3 phosphorylation status during infection, as this may regulate transport activity.

  • Employ conformation-specific antibodies (if available) to detect structural changes upon viral protein binding.

  • Implement multiplexed immunofluorescence to simultaneously visualize CAT3, viral proteins, and markers of cellular stress response.

How can researchers address antibody cross-reactivity issues when studying CAT3 in relation to other cationic amino acid transporters?

Cross-reactivity with other cationic amino acid transporters (particularly CAT1/SLC7A1 and CAT2/SLC7A2) presents a significant challenge in CAT3-focused research due to structural similarities within this family:

Advanced Cross-reactivity Assessment and Mitigation Strategies:

  • Comprehensive Specificity Profiling:

    • Test antibody reactivity against recombinant CAT1, CAT2, and CAT3 proteins in parallel.

    • Perform epitope mapping to identify CAT3-unique versus conserved recognition sites.

    • Generate a cross-reactivity matrix documenting antibody performance across all CAT family members.

  • Genetic Validation Approaches:

    • Use CRISPR/Cas9-mediated knockout of each CAT family member individually and in combination.

    • Implement selective knockdown experiments with siRNAs targeting unique regions of each transporter.

    • Create cell lines expressing epitope-tagged versions of each CAT protein for unambiguous identification.

  • Differential Expression Analysis:

    • Leverage tissue specificity patterns—CAT3 is highly expressed in thymus, uterus, and testis , whereas CAT1 is more ubiquitous.

    • Use quantitative PCR to confirm expression levels of each CAT family member in your experimental system.

    • Perform western blots with carefully optimized conditions to separate CAT isoforms based on molecular weight differences.

  • Technical Refinements for Enhanced Specificity:

    • Implement immunodepletion strategies where antibodies are pre-adsorbed against recombinant non-target CAT proteins.

    • Optimize antibody concentration through titration experiments to identify conditions that maximize specificity.

    • Employ competitive binding assays with increasing concentrations of immunizing peptide to demonstrate binding specificity.

Decision Matrix for Antibody Selection Based on Application:

ApplicationCritical Selection CriteriaRecommended Validation
Western BlotResolution of molecular weight differencesInclude positive controls for each CAT member
ImmunoprecipitationMinimal cross-reactivity in solutionMass spectrometry verification of pulled-down proteins
ImmunohistochemistryDistinct tissue staining patternsComparison with mRNA expression data from public databases
Flow CytometrySignal-to-noise ratio in relevant cell typesFluorescence-minus-one (FMO) controls with CAT knockout cells
ChIP/RIPSpecificity under crosslinking conditionsSequencing verification of enriched regions

Experimental Design for Differential Analysis:

  • For functional studies, use selective inhibitors in combination with genetic approaches to dissect the contribution of individual CAT members.

  • Consider compensatory upregulation of CAT1/CAT2 in CAT3-deficient models when interpreting results .

  • Implement parallel detection of all CAT family members when feasible to provide context for CAT3-specific findings.

  • When absolute specificity cannot be guaranteed, triangulate results using orthogonal methods not dependent on antibody specificity.

What are the most reliable protocols for analyzing CAT3 degradation through the ubiquitin-proteasome pathway?

Investigating CAT3 degradation through the ubiquitin-proteasome pathway requires specialized techniques to capture the dynamic nature of this process :

Comprehensive Protocol Framework:

  • Ubiquitination Analysis:

    • Perform denaturing immunoprecipitation using CAT3 antibodies followed by ubiquitin immunoblotting.

    • Include a panel of proteasome inhibitors (MG132, bortezomib, lactacystin) at optimized concentrations and treatment durations.

    • Use tandem ubiquitin binding entities (TUBEs) to enrich for ubiquitinated proteins before CAT3 detection.

    • Implement ubiquitin remnant profiling (K-ε-GG) with mass spectrometry to identify specific ubiquitination sites on CAT3.

  • Protein Turnover Assessment:

    • Conduct cycloheximide chase experiments comparing CAT3 degradation kinetics in normal versus proteasome-inhibited conditions.

    • Employ pulse-chase labeling with stable isotopes (SILAC) for quantitative degradation rate determination.

    • Implement fluorescent timer fusion proteins that change color based on protein age to visualize CAT3 turnover in situ.

    • Compare degradation rates in wild-type versus ubiquitin mutant (K48R, K63R) expressing cells to determine linkage specificity.

  • E3 Ligase Identification:

    • Perform CRISPR screens targeting E3 ligases to identify those affecting CAT3 stability.

    • Use proximity-dependent biotinylation (BioID) with CAT3 as bait to identify physically associated E3 ligases.

    • Conduct in vitro ubiquitination assays with purified components to confirm direct E3 ligase activity.

  • Integrated Analysis in Viral Infection Context:

    • Compare CAT3 ubiquitination and degradation kinetics in uninfected versus CMV-infected samples .

    • Assess the impact of viral protein (particularly 2b protein) expression on CAT3 stability.

    • Investigate whether viral infection alters the complement of E3 ligases targeting CAT3.

Experimental Design Considerations and Controls:

Experimental AspectCritical ControlsTechnical Considerations
Proteasome inhibitionInclude both reversible (MG132) and irreversible (lactacystin) inhibitorsOptimize concentration to avoid cytotoxicity
Ubiquitin detectionUse linkage-specific antibodies (K48, K63)Denaturing conditions essential to disrupt non-covalent interactions
Degradation kineticsInclude stable proteins as internal controlsAccount for indirect effects on protein synthesis
E3 ligase validationSingle and combinatorial knockdownsConsider redundancy among E3 ligases
Infection modelsInclude viral mutants lacking 2b proteinControl for general effects of cellular stress on proteostasis

Advanced Quantification Approaches:

  • Implement targeted proteomics (PRM/MRM) for precise quantification of ubiquitinated CAT3 peptides.

  • Use fluorescence correlation spectroscopy to measure diffusion properties of CAT3, which change upon ubiquitination.

  • Apply mathematical modeling to deconvolute synthesis, ubiquitination, and degradation rates from experimental data.

  • Integrate transcriptomic and proteomic data to distinguish between transcriptional and post-translational regulation of CAT3 levels.

How can researchers address false-negative results when detecting low abundance CAT3 in tissues with limited expression?

Detecting low-abundance CAT3 in tissues with limited expression presents significant technical challenges that require specialized approaches:

Comprehensive Troubleshooting Strategy:

  • Sample Preparation Optimization:

    • Implement membrane protein enrichment protocols such as two-phase partitioning or sucrose gradient fractionation.

    • Use smaller tissue sections or laser capture microdissection to focus on regions with higher CAT3 expression.

    • Consider sample pooling from multiple specimens to reach detection thresholds.

    • Optimize protein extraction buffers with stronger detergents (1-2% SDS) for complete solubilization.

  • Signal Amplification Approaches:

    • Employ tyramide signal amplification (TSA) for immunohistochemical applications, which can increase sensitivity 10-100 fold.

    • Use high-sensitivity detection systems such as SuperSignal West Femto or ECL Prime for western blots.

    • Implement biotin-streptavidin amplification systems with careful blocking of endogenous biotin.

    • For flow cytometry, use fluorescent tertiary antibodies or branched DNA technology for signal enhancement.

  • Instrumentation and Detection Optimization:

    • Utilize high-sensitivity CCD cameras with extended exposure times for western blot imaging.

    • Employ photomultiplier tube (PMT) detectors with optimized gain settings for confocal microscopy.

    • Consider spectral unmixing to separate CAT3 signal from tissue autofluorescence.

    • Use digital droplet PCR (ddPCR) as a complementary approach to confirm expression at the mRNA level.

  • Antibody Selection and Usage Optimization:

    • Test multiple antibody clones targeting different CAT3 epitopes.

    • Optimize antibody concentration through systematic titration experiments.

    • Extend primary antibody incubation times (overnight at 4°C or 48-72 hours for IHC).

    • Consider using antibody cocktails targeting multiple epitopes simultaneously.

Decision Tree for Troubleshooting Low CAT3 Signal:

ObservationPotential CausesRecommended Actions
No signal in any tissueAntibody failure or technical issueTest antibody on positive control (thymus, uterus, testis) ; check secondary antibody
Signal in controls but not in test tissueLow expression or epitope inaccessibilityTry alternative antibody; increase sample concentration; use signal amplification
Weak, inconsistent signalBorderline expression levelsOptimize all detection parameters; consider pooling samples
Signal at unexpected molecular weightPost-translational modifications or degradationUse deglycosylation enzymes ; add protease and proteasome inhibitors
High background with weak specific signalNon-specific bindingOptimize blocking; increase washing stringency; try alternative antibody

Validation Approaches for Low Abundance Detection:

  • Implement parallel detection of CAT3 mRNA using in situ hybridization or RNAscope to confirm expression in tissues with weak antibody signal.

  • Use transgenic overexpression models as positive controls to establish the detection limit of your experimental system.

  • Perform spiking experiments with recombinant CAT3 to establish a standard curve for quantification in low-expression samples.

  • Consider functional transport assays using radiolabeled amino acids as an indirect measure of CAT3 presence.

What are the optimal experimental designs for studying CAT3's role in oxidative stress responses during viral infection?

Investigating CAT3's role in oxidative stress responses during viral infection requires carefully designed experiments that integrate multiple analytical approaches :

Comprehensive Experimental Design Framework:

  • Model System Selection and Validation:

    • Compare wild-type, CAT3-knockout, and CAT3-overexpressing systems in parallel .

    • Include viral mutants lacking specific components (e.g., 2b protein) to dissect interaction mechanisms .

    • Validate model systems by confirming CAT3 expression levels via western blot and qPCR.

    • Establish baseline oxidative status parameters prior to infection.

  • Infection Protocol Optimization:

    • Implement time-course experiments with multiple sampling points (early, peak, and late infection phases).

    • Use multiple virus concentrations (MOI) to establish dose-dependent effects.

    • Include appropriate mock-infected controls with identical treatment except for virus.

    • Consider comparing different viral strains to identify CAT3-specific versus general infection responses.

  • Oxidative Stress Measurement Strategy:

    • Implement a multi-parameter approach combining direct H₂O₂ measurements, oxidative damage markers, and antioxidant system status.

    • Perform real-time monitoring using genetically encoded redox sensors (roGFP, HyPer) in live cells.

    • Conduct subcellular fractionation to assess compartment-specific oxidative changes.

    • Use fluorescence lifetime imaging (FLIM) to detect subtle changes in redox sensor response.

  • CAT3-Specific Analysis:

    • Track CAT3 protein levels, subcellular localization, and post-translational modifications throughout infection.

    • Assess CAT3-viral protein interaction dynamics using proximity ligation assay (PLA) or FRET.

    • Monitor CAT3 degradation kinetics with and without proteasome inhibitors .

    • Evaluate CAT3 transporter activity using radiolabeled amino acid uptake assays .

Integrated Analysis Approach:

ParameterMethodsControls and Considerations
H₂O₂ LevelsAmplex Red assay; HyPer fluorescent protein; PeroxyGreen probesInclude catalase treatment controls; account for probe photostability
Oxidative Damage8-OHdG (DNA); protein carbonylation; lipid peroxidation (TBARS, 4-HNE)Compare wild-type vs. CAT3-knockout ; include antioxidant treatment arms
Antioxidant SystemsGSH/GSSG ratio; SOD, catalase, GPx activities; Nrf2 pathway activationMeasure both protein levels and activities; assess compensatory responses
Cell Death ParametersApoptosis vs. necrosis discrimination; ferroptosis markersCorrelate with oxidative parameters; compare to CAT3 expression levels
Viral ReplicationPlaque assays; viral protein/RNA quantificationCompare replication kinetics across genotypes and oxidative conditions

Advanced Analytical Approaches:

  • Implement redox proteomics to identify oxidation-sensitive proteins in CAT3-dependent and independent pathways.

  • Use metabolomics to track changes in redox-related metabolites (GSH, NADPH, ascorbate) during infection.

  • Perform ChIP-seq for redox-sensitive transcription factors (Nrf2, NF-κB) to link oxidative changes to transcriptional responses.

  • Apply spatial transcriptomics to correlate local CAT3 expression with oxidative stress signatures in infected tissues.

  • Implement systems biology approaches to model the interplay between CAT3, viral proteins, and oxidative stress networks.

How can researchers develop optimal immunoprecipitation protocols for studying CAT3 interactions with viral and host proteins?

Developing effective immunoprecipitation (IP) protocols for studying CAT3 interactions with viral and host proteins requires careful optimization due to CAT3's membrane localization and the potentially transient nature of its interactions :

Comprehensive IP Protocol Development Framework:

  • Antibody Selection and Validation:

    • Test multiple anti-CAT3 antibodies targeting different epitopes.

    • Validate each antibody's IP efficiency using western blot detection of CAT3 in input versus immunoprecipitated fractions.

    • Confirm that selected antibodies do not interfere with protein-protein interaction interfaces.

    • Consider epitope-tagged CAT3 constructs (HA, FLAG, etc.) as alternatives when direct CAT3 antibodies show limitations.

  • Lysis and Solubilization Optimization:

    • Systematically test a panel of detergents (NP-40, Triton X-100, digitonin, CHAPS) at different concentrations.

    • Optimize detergent:protein ratios to ensure complete solubilization without disrupting interactions.

    • For stronger interactions, consider more stringent buffers (RIPA); for weaker interactions, use milder conditions (0.3% CHAPS).

    • Include protease, phosphatase, and proteasome inhibitors to preserve interaction status .

  • Interaction Stabilization Strategies:

    • Implement chemical crosslinking (DSP, formaldehyde) with optimized concentration and time parameters.

    • Use proximity-dependent labeling (BioID, APEX) as complementary approaches for capturing transient interactions.

    • Consider conducting IPs at different temperatures (4°C, room temperature) to balance interaction preservation versus non-specific binding.

    • For RNA-dependent interactions, include RNase inhibitors or conduct parallel experiments with RNase treatment.

  • Control Strategy Implementation:

    • Include multiple control IPs: isotype control antibodies, pre-immune serum, and IPs from CAT3-knockout samples .

    • Implement competition controls with immunizing peptides at increasing concentrations.

    • For viral interaction studies, include uninfected samples and infections with mutant viruses lacking interaction partners .

    • Consider reverse IPs (pull-down using antibodies against suspected interaction partners) for validation.

Protocol Optimization Decision Matrix:

ParameterOptimization RangeEvaluation MetricsConsiderations
Cell Lysis ConditionsDetergent type (0.3-1%); buffer composition; incubation timeCAT3 recovery in soluble fraction; preservation of known interactionsBalance between solubilization efficiency and maintenance of interactions
Antibody Amount1-10 μg per mg of proteinPull-down efficiency; backgroundExcess antibody can increase non-specific binding
Incubation Time2 hours to overnightYield of target and interacting proteinsLonger incubations may increase yield but also background
Washing StringencyNumber of washes; salt concentration (150-500 mM)Specific vs. non-specific binding ratioMore stringent washing reduces false positives but may lose weak interactions
Elution MethodNative (peptide competition) vs. denaturing (SDS, boiling)Recovery efficiency; compatibility with downstream analysisChoose based on intended subsequent analysis

Advanced Analytical Strategies:

  • Implement sequential IPs to isolate specific subcomplexes (tandem affinity purification approach).

  • Use isotope labeling (SILAC) combined with IP for quantitative comparison of interaction dynamics across conditions.

  • Couple IP with mass spectrometry for unbiased identification of the complete CAT3 interactome.

  • Implement proximity-dependent biotin identification (BioID) with CAT3-BioID fusion proteins as a complementary approach.

  • For viral protein interactions, consider in situ approaches such as PLA to validate interactions in their native cellular context.

How can emerging antibody engineering technologies be applied to develop more specific and sensitive CAT3 detection tools?

Emerging antibody engineering technologies offer significant opportunities to develop next-generation CAT3 detection tools with enhanced specificity, sensitivity, and functionality:

Advanced Antibody Engineering Approaches:

  • Computational Design and Screening:

    • Implement machine learning algorithms to predict optimal CAT3 epitopes based on accessibility and uniqueness .

    • Use structural modeling to design antibodies with enhanced CAT3 specificity versus other CAT family members.

    • Apply in silico affinity maturation to improve sensitivity without compromising specificity .

    • Leverage large antibody sequence datasets for identifying naturally occurring anti-CAT3 sequences with desirable properties .

  • Single-Domain Antibody Development:

    • Generate camelid VHH (nanobodies) or shark VNAR antibodies against CAT3 for enhanced access to conformational epitopes.

    • Optimize nanobodies for specific applications such as super-resolution imaging or intracellular expression.

    • Create nanobody libraries against intact membrane-embedded CAT3 to target native conformations.

    • Develop biparatopic nanobodies targeting two different CAT3 epitopes simultaneously for enhanced specificity and avidity.

  • Antibody Fragment and Alternative Scaffold Technologies:

    • Generate Fab, scFv, or diabody formats for improved tissue penetration in imaging applications.

    • Explore non-antibody scaffolds (DARPins, Affibodies, Anticalins) for CAT3 recognition with novel binding properties.

    • Create recombinant fusion proteins combining CAT3-specific binding domains with detection or functional moieties.

    • Develop aptamer alternatives to complement antibody-based detection systems.

  • Multimodal and Switchable Antibody Technologies:

    • Design pH- or redox-sensitive antibodies that bind CAT3 only under specific cellular conditions.

    • Develop bispecific antibodies that simultaneously target CAT3 and interaction partners for in situ complex detection.

    • Create antibody-based molecular sensors that change conformation or emit signals upon CAT3 binding.

    • Implement optogenetic or chemogenetic control elements for temporally regulated CAT3 detection or modulation.

Application-Specific Engineering Considerations:

ApplicationEngineering ApproachAnticipated BenefitsDevelopment Considerations
Live-Cell ImagingFluorescent nanobodies; Fab fragments with site-specific dye conjugationMinimal interference with CAT3 function; improved signal-to-noiseMaintain native CAT3 distribution and transport activity
Super-Resolution MicroscopySmall binding domains with optimal fluorophore positioningEnhanced spatial resolution of CAT3 organization in membraneStrategic positioning of fluorophores relative to binding site
Intracellular DetectionCell-penetrating antibody fragments; genetically encoded intrabodiesDirect monitoring of CAT3 in living cellsSelection for stability in reducing intracellular environment
Multiplex DetectionOrthogonal labeling strategies; spectrally distinct reportersSimultaneous visualization of CAT3 with interaction partnersMinimize crosstalk between detection channels
Proteomic AnalysisMass-spec compatible conjugates; cleavable crosslinkersEnhanced identification of CAT3 complexesDesign for compatibility with sample preparation requirements

Implementation Strategy and Validation Framework:

  • Establish clear benchmarks against conventional antibodies for sensitivity, specificity, and reproducibility.

  • Validate engineered antibodies across multiple platforms (western blot, IP, IHC, flow cytometry) to ensure versatility.

  • Confirm that binding of engineered formats does not interfere with CAT3 transport function using amino acid uptake assays .

  • Perform comprehensive cross-reactivity testing against all CAT family members and structurally related transporters.

  • Validate performance in relevant biological contexts, particularly viral infection models where CAT3 function is modulated .

What are the most promising approaches for developing antibodies that can distinguish between different functional states of CAT3?

Developing antibodies that can distinguish between different functional states of CAT3 (active/inactive, ligand-bound/unbound, different PTM states) represents an important frontier in CAT3 research:

State-Specific Antibody Development Strategies:

  • Conformational State Recognition:

    • Design immunization strategies using stabilized CAT3 in specific conformational states (e.g., using conformation-specific inhibitors or activators).

    • Implement phage display selections with alternating positive and negative selection rounds against different CAT3 conformations .

    • Use structural information to identify epitopes that undergo significant conformational changes during transport cycles.

    • Develop llama nanobodies which excel at recognizing conformational epitopes due to their long CDR3 regions .

  • PTM-State Specific Antibodies:

    • Generate antibodies against synthetic peptides containing specific PTMs known to regulate CAT3 (phosphorylation, ubiquitination, glycosylation) .

    • Implement rigorous screening protocols to confirm specificity for the modified versus unmodified forms.

    • Develop bispecific antibodies that simultaneously recognize the CAT3 backbone and specific modifications.

    • Create antibodies that specifically recognize proteolytic processing states or degradation intermediates of CAT3 .

  • Ligand/Interaction-State Specific Recognition:

    • Design selection strategies to isolate antibodies that preferentially bind CAT3 in complex with specific transported amino acids .

    • Develop antibodies that specifically recognize CAT3-viral protein complexes versus free CAT3 .

    • Create antibodies that distinguish between membrane-localized versus internalized CAT3.

    • Implement conditional binding domains that only recognize CAT3 under specific microenvironmental conditions.

  • Activity-Based Recognition Approaches:

    • Develop antibodies against CAT3 trapped in transport-competent versus transport-incompetent states.

    • Create sensors based on antibody fragments that report on CAT3 transport activity through conformational coupling.

    • Engineer antibody-based proximity sensors that report on CAT3 interactions with key regulatory proteins.

    • Implement activity-based protein profiling approaches to label and detect only functionally active CAT3.

Validation Framework for State-Specific Antibodies:

Functional StateValidation ApproachesControl ConditionsApplications
Transport-active vs. inactiveAmino acid uptake correlation; conformational mutantsTransport inhibitors; activating conditionsMonitoring CAT3 functional status in disease models
Phosphorylated vs. non-phosphorylatedPhosphatase treatment; site-directed mutagenesisKinase inhibitors/activatorsTracking regulatory mechanisms
Ubiquitinated vs. non-ubiquitinatedDeubiquitinase treatment; proteasome inhibitionUbiquitin pathway modulatorsStudying degradation dynamics
Viral protein-bound vs. unboundIn vitro complex formation; infection modelsMutant viral proteinsInvestigating infection mechanisms
Glycosylated vs. non-glycosylatedGlycosidase treatment; glycosylation site mutantsGlycosylation inhibitorsAssessing maturation status

Experimental Design Considerations:

  • Implement multiplexed detection systems that simultaneously monitor multiple CAT3 states using differentially labeled state-specific antibodies.

  • Design time-course experiments to track transitions between functional states during physiological processes or pathological conditions.

  • Combine state-specific antibodies with functional assays to correlate specific states with CAT3 activity levels.

  • Integrate advanced imaging approaches such as FRET, FLIM, or single-molecule tracking to monitor state transitions in real-time.

  • Develop quantitative assays that provide precise measurements of the proportion of CAT3 in each functional state.

How might high-throughput antibody discovery technologies be leveraged to develop comprehensive CAT3 antibody toolkits for diverse research applications?

High-throughput antibody discovery technologies offer unprecedented opportunities to develop comprehensive CAT3 antibody toolkits spanning multiple applications, epitopes, and detection modalities:

Strategic Implementation of High-Throughput Discovery Platforms:

  • Next-Generation Phage/Yeast Display Technologies:

    • Implement deep mutational scanning of antibody libraries against CAT3 to identify optimal binding sequences .

    • Apply orthogonal selection pressures to isolate application-specific binders (e.g., selecting for heat stability, detergent compatibility).

    • Use microfluidic sorting platforms to rapidly screen millions of potential CAT3 binders based on affinity and specificity .

    • Implement sequential positive-negative selection cycles to ensure specificity against other CAT family members.

  • Single B-Cell Screening and Microfluidic Approaches:

    • Isolate and screen antibody-secreting cells (ASCs) from immunized animals using microfluidic platforms .

    • Implement rapid screening of natural antibody repertoires against multiple CAT3 epitopes simultaneously.

    • Apply droplet microfluidics to achieve ultra-high-throughput screening of antibody-secreting cells with minimal sample requirements .

    • Integrate functional assays directly into the screening workflow to identify antibodies that modulate CAT3 activity.

  • Synthetic Library and Rational Design Approaches:

    • Leverage computational antibody design to create focused libraries targeting specific CAT3 epitopes .

    • Apply machine learning algorithms to predict optimal CAT3-binding sequences based on existing antibody datasets .

    • Implement massively parallel DNA synthesis to create diverse libraries targeting predicted CAT3 epitope regions.

    • Use structure-guided design to develop antibodies with enhanced specificity for CAT3 versus other CAT family members.

  • Integrative Multi-Platform Approaches:

    • Combine multiple discovery platforms to leverage the strengths of each approach.

    • Implement cross-validation between platforms to ensure robust identification of high-quality CAT3 binders.

    • Develop pipeline approaches where initial hits from high-throughput screens undergo stepwise optimization and validation.

    • Create systematic epitope binning strategies to ensure comprehensive coverage of the CAT3 structure.

Comprehensive Toolkit Development Strategy:

Application CategoryAntibody RequirementsDiscovery/Selection StrategyValidation Framework
Detection/QuantificationHigh specificity and sensitivity; compatibility with multiple platformsMulti-parameter selection for affinity, specificity, and stabilityCross-platform performance testing; limit of detection determination
Functional ModulationAbility to inhibit or activate CAT3 transportSelection with functional readouts (transport assays) Dose-response assessment; specificity of functional effect
ImagingMinimal background; photostability; site-specific labelingSelection under conditions mimicking imaging applicationsSignal-to-noise optimization; co-localization with established markers
Interaction StudiesNon-interference with protein-protein interfacesEpitope mapping and selection away from interaction sitesValidation in multiple interaction assay formats
PTM DetectionAbsolute specificity for modified formsSophisticated negative selection against unmodified CAT3Verification with enzymes that add/remove modifications

Implementation and Data Management Framework:

  • Create a centralized database of CAT3 epitopes, antibody sequences, and performance characteristics across applications .

  • Implement standardized validation protocols to enable direct comparison between antibodies.

  • Develop application-specific panels where multiple antibodies can be used in combination for enhanced performance.

  • Establish a decision tree to guide researchers in selecting optimal antibodies for specific experimental questions.

  • Create an accessible repository of recombinant antibody expression constructs to facilitate widespread adoption and further optimization.

Broader Research Community Integration:

  • Establish collaborative networks for systematic validation across multiple laboratories and applications.

  • Implement open science principles with detailed protocols and validation data made publicly available.

  • Develop standardized reporting formats for CAT3 antibody characteristics to enable meaningful comparison across studies.

  • Create educational resources to guide appropriate selection and implementation of CAT3-specific antibody tools.

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.