ADS2 Antibody

Shipped with Ice Packs
In Stock

Description

Potential Confusion with Established Antibody Targets

Two antibodies with similar nomenclature demonstrate significant scientific importance:

Table 1: Comparison of Related Antibodies

ParameterADD2 Antibody (Beta-Adducin) Anti-DSG2 Antibody
Target ProteinBeta-adducin (ADD2)Desmoglein-2 (DSG2)
Host SpeciesRabbit polyclonalHuman autoantibodies
ApplicationsWB/IF/ELISAAutoimmune disease diagnostics
Clinical ContextCytoskeletal regulationArrhythmogenic cardiomyopathy
Key ReferencesSTJ91491 PMC11594951 , PMC9405509

Detailed Characterization of ADD2 Antibody

From the identified anti-ADD2 antibody (STJ91491) :

  • Immunogen: Synthetic peptide (amino acids 471-520 of human ADD2)

  • Concentration: 1 mg/mL

  • Cross-Reactivity: Human, Mouse, Rat

  • Storage: -20°C with sodium azide preservative

Functional Role:
Binds to β-adducin, a membrane-cytoskeleton protein critical for:

  • Spectrin-actin network assembly

  • Erythrocyte membrane stabilization via SLC2A1/GLUT1 interaction

  • Calcium-regulated signaling through calmodulin binding

Anti-DSG2 Antibody Findings

Recent studies demonstrate elevated DSG2 autoantibodies in:

  • 56% of arrhythmogenic right ventricular cardiomyopathy (ARVC) cases

  • 29.3% of post-COVID-19 patients (persisting ≥9 months post-infection)

Table 2: Anti-DSG2 Clinical Correlates

Patient CohortPrevalenceMedian TiterPathological Association
ARVC56%18.7 U/LVentricular arrhythmias
Post-COVID-19 (6mo)29.3%19.0 ODSustained cardiac autoimmunity
Healthy Controls<5%2.1 ODNone

Technical Validation Methods

For antibodies targeting similar epitopes:

ADA Testing Protocols :

  1. Screening ELISA (cutoff: 3x background)

  2. Confirmatory competitive inhibition assay

  3. Neutralization testing (≥50% signal reduction)

Key Quality Metrics:

  • Drug-to-antibody ratio (DAR): 3.5-4.0 optimal

  • ADA incidence thresholds:

    • Low risk: <2% (e.g., omalizumab )

    • High risk: >8% (e.g., benralizumab )

Research Recommendations

To resolve the ADS2 ambiguity:

  1. Verify nomenclature with IUPAC/IUIS databases

  2. Perform BLAST alignment against:

    • Adducin family (ADD1-3)

    • Desmoglein isoforms (DSG1-4)

  3. Screen for potential cross-reactivity in existing ELISA panels

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ADS2; At2g31360; T28P16.15; Delta-9 acyl-lipid desaturase 2
Target Names
ADS2
Uniprot No.

Target Background

Function
ADS2 is a crucial enzyme involved in the delta-9 desaturation of fatty acids.
Gene References Into Functions
  1. Research suggests that ADS2 plays a critical role in adjusting the acyl composition of organelle membrane lipid composition in response to cold stress. PMID: 23585650
  2. Studies have shown that AtADS2 is involved in the biosynthesis of glycerolipid and sphingolipids within the seed. PMID: 23175755
Database Links

KEGG: ath:AT2G31360

STRING: 3702.AT2G31360.1

UniGene: At.14455

Protein Families
Fatty acid desaturase type 1 family
Subcellular Location
Endoplasmic reticulum membrane; Multi-pass membrane protein.
Tissue Specificity
Strongly expressed in flowers, roots, leaves, seedpods, and inflorescence meristems.

Q&A

What are the recommended detection methods for ADS2 antibodies in different sample types?

Detection of ADS2 antibodies requires selecting appropriate methodologies based on sample type. For plasma and mucosal samples (such as saliva), enzyme-linked immunosorbent assays (ELISAs) are commonly employed to quantify total immunoglobulin levels against the target antigen .

When working with plasma samples, titration curves against specific antigens provide robust quantification. In contrast, saliva samples typically yield significantly lower antibody concentrations but maintain strong correlation with plasma levels for total immunoglobulin against tested antigens . The detection hierarchy typically follows:

  • Initial screening: Total antibody assessment via ELISA

  • Isotype determination: IgG1, IgM, IgA1 quantification

  • Functional assessment: For antibodies with neutralizing capacity

In research settings, Ligand Binding Assays (LBAs) or hybrid LC-MS/MS approaches provide complementary data for comprehensive antibody characterization . These methods allow for more detailed binding specificity analysis and can distinguish between different antibody isotypes and subtypes.

How should samples be collected and handled to ensure optimal ADS2 antibody detection?

Proper sample collection and handling are critical for accurate ADS2 antibody detection:

Plasma Collection:

  • Collect blood in EDTA or heparin tubes

  • Process within 2-4 hours of collection

  • Centrifuge at 1500-2000g for 10 minutes at room temperature

  • Aliquot plasma to avoid freeze-thaw cycles and store at -80°C

Saliva Collection:

  • Collect unstimulated saliva in sterile containers

  • Maintain cold chain (2-8°C) during transportation

  • Centrifuge at 2500g for 10 minutes to remove cellular debris

  • Filter through 0.45μm filters if necessary

  • Store aliquots at -80°C

Studies have shown that antibody detection remains reliable in properly stored samples for up to 12 months at -80°C . When comparing across sample types, standardized collection protocols are essential as differences in collection methodology can significantly impact quantification results.

What controls should be included in ADS2 antibody detection assays?

A comprehensive control strategy is essential for reliable ADS2 antibody detection:

Essential Controls for Antibody Detection Assays:

Control TypePurposeImplementation
Positive ControlValidates assay performanceKnown ADS2 antibody-positive sample
Negative ControlEstablishes baselineSample from unexposed individual
Isotype ControlAssesses specificityNon-specific antibody of same isotype
Matrix ControlEvaluates matrix effectsBuffer with similar components to test sample
Cut-off ControlDetermines positivity thresholdSamples near detection limit

Additionally, for multi-tiered testing schemes, bovine serum albumin (BSA) can serve as an effective negative control antigen to assess non-specific binding . For experiments involving multiple timepoints, longitudinal controls should be included to account for inter-assay variability.

How do ADS2 antibody levels differ between plasma and mucosal samples?

Significant differences exist in ADS2 antibody levels and isotype distribution between plasma and mucosal samples:

  • Plasma samples typically show substantially higher antibody concentrations compared to saliva samples, though strong correlations exist between levels in both matrices

  • IgG1 responses predominate in both plasma and saliva samples

  • Lower prevalence of IgM and IgA1 antibodies is typically observed in saliva compared to plasma

  • Despite lower absolute concentrations, mucosal antibodies represent the frontline defense against respiratory pathogens

Research has demonstrated that while absolute concentrations differ, relative changes in antibody levels often follow similar patterns in both sample types, making saliva a potentially valuable non-invasive alternative for monitoring antibody responses in longitudinal studies .

What is the optimal experimental design for evaluating ADS2 antibody kinetics in longitudinal studies?

Designing longitudinal studies to evaluate ADS2 antibody kinetics requires careful consideration of sampling strategy, analysis methods, and potential confounding factors:

Recommended Sampling Strategy:

Study PhaseSampling FrequencyKey Parameters to Measure
BaselineSingle timepointPre-exposure antibody levels, isotype distribution
Initial Response1-2 weeks post-exposureAntibody induction, isotype switching
Peak Response2-6 weeks post-exposureMaximum antibody titers, functional activity
Decay PhaseMonthly for 6+ monthsAntibody persistence, affinity maturation
Memory Phase6-12 months post-exposureLong-term antibody maintenance

For comprehensive analysis, implement a multi-parametric approach that assesses:

  • Total antibody concentration using LBA or hybrid LC-MS/MS methods

  • Functional activity through appropriate bioassays

  • Isotype distribution via isotype-specific detection reagents

  • Epitope specificity through competition assays

Statistical power calculations should account for expected inter-individual variability (typically 30-50% for antibody responses) and anticipated dropout rates in longitudinal designs .

How can immunogenicity of ADS2 antibodies be systematically analyzed and reported?

Systematic analysis of ADS2 antibody immunogenicity requires a structured, multi-tiered approach:

Tiered Analysis Strategy:

  • Screening Assay: Initial detection of anti-ADS2 antibodies using sensitive methods

  • Confirmatory Assay: Validation of positive screening results through competitive inhibition

  • Characterization: Determination of antibody isotypes, titers, and neutralizing capacity

  • Impact Assessment: Evaluation of effects on pharmacokinetics, pharmacodynamics, and clinical outcomes

Key Derived Parameters for Comprehensive Analysis:

  • Treatment-induced antibody response (≥4-fold increase in post-baseline titer)

  • Post-baseline positive antibody incidence

  • Time to onset of first antibody detection

  • Persistently positive antibody presence (positive at ≥2 timepoints with ≥16 weeks between first and last positive)

  • Transiently positive antibody response (at least one positive result but not meeting persistence criteria)

  • Neutralizing antibody incidence

These parameters should be analyzed in relation to clinical outcomes and pharmacokinetic data to determine the impact of immunogenicity on therapeutic efficacy and safety.

What approaches are recommended for resolving contradictory ADS2 antibody data from different detection methods?

When faced with contradictory ADS2 antibody results from different detection methods, a systematic troubleshooting approach is essential:

  • Evaluate method sensitivity and specificity:

    • Different methods have varying detection limits and cross-reactivity profiles

    • Compare lower limits of quantification and upper limits of linearity

    • Assess potential matrix effects

  • Consider epitope accessibility:

    • Conformational changes in antigens can mask epitopes in certain assay formats

    • Solution-phase versus solid-phase binding may yield different results

    • Denaturation during sample processing may alter epitope recognition

  • Implement orthogonal testing strategy:

    • Combine binding assays (ELISA, LBA) with functional tests

    • Apply both ligand binding and LC-MS/MS approaches

    • Utilize epitope mapping to identify recognition sites

  • Conduct method comparison studies:

    • Analyze a subset of samples with all methods in parallel

    • Determine Bland-Altman plots to assess systematic bias

    • Calculate correlation coefficients and concordance rates

When method discrepancies persist, results from neutralization assays often provide the most clinically relevant information regarding antibody functionality .

How does the presence of ADS2 antibodies impact pharmacokinetic profiles in research models?

The presence of ADS2 antibodies can significantly alter pharmacokinetic profiles through multiple mechanisms:

Impacts on Key PK Parameters:

ParameterEffect of Non-Neutralizing AntibodiesEffect of Neutralizing Antibodies
CmaxModerate reductionSignificant reduction
AUCModerate reductionMarked reduction (>50%)
Half-lifeVariable effectsSubstantially shortened
ClearanceModerately increasedDramatically increased
Volume of distributionGenerally unchangedGenerally unchanged

The binding characteristics of anti-drug antibodies determine their impact. Non-neutralizing antibodies that bind to non-active regions of the therapeutic may have minimal effects on maximum concentration (Cmax), while neutralizing antibodies that bind to active sites substantially increase elimination and dramatically lower Cmax .

The clinical impact model demonstrates that subjects without anti-drug antibodies maintain significantly higher drug concentrations compared to those with antibody titers. This relationship is particularly important when interpreting concentration-time curves and assessing drug efficacy in research models .

What design of experiments (DOE) approach is recommended for optimizing ADS2 antibody detection assays?

Implementing Design of Experiments (DOE) methodology for optimizing ADS2 antibody detection assays allows for systematic evaluation of multiple parameters simultaneously:

Critical Parameters for DOE Optimization:

  • Coating antigen concentration/density

  • Sample dilution factors

  • Incubation times and temperatures

  • Detection antibody concentration

  • Blocking reagent composition

  • Wash buffer formulation

Recommended DOE Approach:

  • Begin with fractional factorial design to screen 6-8 parameters

  • Identify significant factors using analysis of variance (ANOVA)

  • Implement response surface methodology on critical factors

  • Establish design space for robust assay performance

  • Validate optimal conditions with independent samples

This systematic approach offers several advantages over one-factor-at-a-time optimization, including identification of parameter interactions, reduced experimental runs, and enhanced statistical power .

For multi-tiered testing schemes, DOE should be applied to each tier individually, with optimized conditions from earlier tiers informing the starting points for subsequent tier optimization.

What are the current standardized methods for distinguishing between neutralizing and non-neutralizing ADS2 antibodies?

Distinguishing between neutralizing and non-neutralizing ADS2 antibodies requires specialized assays that evaluate functional impacts:

Competitive Binding Assays:
These assess whether antibodies interfere with therapeutic binding to its target. While relatively simple to implement, they may not fully predict functional neutralization.

Cell-Based Neutralization Assays:
These provide direct evidence of functional neutralization by measuring inhibition of biological activity. Critical considerations include:

  • Selection of appropriate cell line expressing relevant receptors

  • Development of validated readouts (reporter genes, viability, etc.)

  • Inclusion of reference standards for normalization

  • Determination of neutralizing titer cutpoints

Hybrid Approaches:
For complex research questions, combining binding assays with functional assays provides comprehensive characterization. The immunogenicity testing pyramid progresses from:

  • Initial screening (binding)

  • Confirmatory testing (competitive binding)

  • Neutralization assessment (functional)

  • Characterization (isotyping, epitope mapping)

Non-neutralizing antibodies bind to the therapeutic without affecting target binding, potentially altering drug half-life. In contrast, neutralizing antibodies interact directly with pharmacologically relevant sites, obscuring interactions between the therapeutic and its target .

How can researchers effectively analyze the relationship between ADS2 antibody titers and clinical outcomes?

Analyzing relationships between ADS2 antibody titers and clinical outcomes requires robust statistical approaches and careful data interpretation:

Analytical Framework:

  • Categorization of Antibody Responses:

    • Classify subjects by antibody status (negative, transiently positive, persistently positive)

    • Group by neutralizing capacity (non-neutralizing vs. neutralizing)

    • Stratify by titer levels (low, medium, high)

  • Statistical Methods for Correlation Analysis:

    • Mixed-effects models for longitudinal data

    • Cox proportional hazards for time-to-event outcomes

    • Logistic regression for binary outcomes

    • Propensity score matching to control for confounding factors

  • Pharmacometric Modeling:

    • Develop population PK/PD models incorporating immunogenicity

    • Simulate concentration-effect relationships under various antibody scenarios

    • Perform covariate analysis to identify factors influencing antibody impact

The presence of antibodies can significantly affect drug concentration-time curves, with neutralizing antibodies dramatically lowering Cmax and area under the curve (AUC). This PK alteration typically translates to reduced clinical efficacy and potentially altered safety profiles .

For early-stage clinical development, key questions to address include incidence rates of antibody formation, impact on PK parameters, and correlation with efficacy endpoints. For late-stage development, focus shifts to immunogenicity risk assessment and definition of appropriate risk mitigation strategies .

What sample preparation techniques maximize detection sensitivity for low-concentration ADS2 antibodies?

Optimizing sample preparation is crucial for detecting low-concentration ADS2 antibodies:

Enhanced Sample Preparation Techniques:

TechniquePrincipleApplication
Affinity PurificationEnrichment using immobilized antigensConcentration of target antibodies
ImmunoprecipitationCapture with protein A/GIsolation from complex matrices
ImmunodepletionRemoval of abundant proteinsReduction of background interference
UltracentrifugationSeparation by densityIsolation of immune complexes

For plasma samples, a sequential approach is recommended:

  • Initial dilution in assay buffer containing blocking agents

  • Pre-absorption against non-specific binding surfaces

  • Removal of particulates by centrifugation (10,000g for 10 minutes)

  • Incubation at appropriate temperature (typically 4°C overnight for maximum binding)

For mucosal samples such as saliva, additional steps are necessary:

  • Filtering through 0.45μm filters to remove cellular debris

  • Buffer exchange to standardize pH and ionic strength

  • Concentration using centrifugal filter units with appropriate molecular weight cutoffs

  • Addition of protease inhibitors to prevent degradation

These techniques can improve detection sensitivity by 5-10 fold compared to standard sample processing methods.

How can researchers accurately quantify drug-to-antibody ratios in ADC research involving ADS2 antibodies?

Accurate quantification of drug-to-antibody ratios (DAR) is essential in antibody-drug conjugate research:

Recommended Analytical Methods:

  • UV-Vis Spectroscopy:

    • Utilizes differential absorbance of antibody and drug components

    • Provides average DAR but limited resolution of DAR species

  • Hydrophobic Interaction Chromatography (HIC):

    • Separates conjugates based on hydrophobicity differences

    • Enables visualization of DAR distribution

  • Mass Spectrometry:

    • Native MS for intact ADC analysis

    • Middle-down approaches for subunit analysis

    • Bottom-up approaches for peptide-level characterization

  • Hybrid LC-MS/MS Approaches:

    • Combines chromatographic separation with MS detection

    • Provides comprehensive DAR distribution and heterogeneity assessment

For early-stage ADC development, screening various ADC stabilities assuming leverageable chemistries is recommended. Both ligand binding assays and hybrid LC-MS/MS using commercially available reagents can be employed for "generic assays" to quantify ADCs .

When analyzing data, calculate both average DAR and DAR distribution, as ADCs with identical average DAR may have different distributions that significantly impact efficacy and safety profiles.

What are the critical quality attributes to monitor during stability studies of ADS2 antibodies?

Stability studies for ADS2 antibodies should monitor several critical quality attributes (CQAs) that impact safety and efficacy:

Essential Critical Quality Attributes:

CategoryAttributeAnalytical MethodAcceptance Criteria
Primary StructureAmino acid sequenceLC-MS/MS peptide mapping≥95% sequence coverage
Secondary/Tertiary StructureConformational integrityCircular dichroism, FTIRConsistent spectral profile
Size VariantsAggregationSEC, DLS<5% high molecular weight species
FragmentationCE-SDS, SDS-PAGE<5% fragments
Charge VariantsDeamidationIEX, LC-MSWithin established ranges
OxidationRP-HPLC, LC-MS<5% increase from baseline
GlycosylationGlycan profileHILIC, MSConsistent pattern
Biological ActivityTarget bindingSPR, ELISA≥80% retained activity
Functional activityCell-based assays≥80% retained function

For accelerated stability studies, implement a matrix design examining multiple conditions:

  • Temperature (2-8°C, 25°C, 40°C)

  • Humidity (60%, 75%)

  • Light exposure (protected, ambient light, forced photodegradation)

  • Agitation (static, controlled stress)

Monitor at pre-defined intervals (initial, 1, 3, 6, 9, 12 months) for comprehensive stability profiling. Stability-indicating methods should be validated to detect changes in antibody structure and function over time.

How should researchers design immunogenicity studies for ADS2 antibodies in preclinical models?

Designing robust immunogenicity studies for ADS2 antibodies in preclinical models requires careful planning:

Comprehensive Study Design Elements:

  • Species Selection:

    • Choose species with similar target biology to humans

    • Consider antibody-target homology across species

    • Evaluate cross-reactivity profiles

  • Dosing Strategy:

    • Include multiple dose levels (low, mid, high)

    • Implement both single-dose and repeat-dose arms

    • Consider route of administration relevance

  • Sampling Schedule:

    • Baseline (pre-dose)

    • Early response (7-14 days post-dose)

    • Peak response (28-35 days post-dose)

    • Extended timepoints for persistence evaluation

  • Analysis Plan:

    • Multi-tiered approach (screening, confirmation, characterization)

    • Combined PK/immunogenicity assessments

    • Correlation with pharmacodynamic markers

  • Control Groups:

    • Vehicle control

    • Irrelevant antibody control (same isotype)

    • Positive control (known immunogenic antibody)

The analysis should address key development-stage questions including immunogenicity risk assessment and potential impact on efficacy and safety . Neutralizing antibody assays are particularly important to include alongside total anti-drug antibody assessments.

What statistical approaches are recommended for establishing cut-points in ADS2 antibody immunogenicity assays?

Establishing statistically sound cut-points is crucial for immunogenicity assay reliability:

Recommended Statistical Approaches:

  • Parametric Method:

    • Calculate mean + 1.645 × SD for 5% false positive rate

    • Applicable when data follow normal distribution

    • Grubbs' test for outlier identification

  • Non-parametric Method:

    • Use 95th or 97.5th percentile directly

    • Appropriate for non-normally distributed data

    • More robust to extreme values

  • Cut-point Factor Approach:

    • Calculate ratio of cut-point to negative control

    • Apply this factor to normalize run-to-run variation

    • Useful for floating cut-point implementation

Cut-point Hierarchy:

  • Screening Cut-point: Liberal threshold (5% false positive rate)

  • Confirmatory Cut-point: More stringent (1% false positive rate)

  • Titer Cut-point: Based on signal-to-noise ratio

For multi-tiered ADA testing schemes, each assay requires its own validated cut-point. Cut-points should be established during assay validation using at least a 50-sample negative population and verified periodically during study conduct . Performance of the cut-point should be monitored throughout the study to ensure consistent sensitivity and specificity.

Quick Inquiry

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