ADT4 Antibody

Shipped with Ice Packs
In Stock

Description

ADT-4 Antibody in Alzheimer’s Disease Research

ADT-4 (Alzheimer’s Disease Tau-4) is a single-chain variable fragment (scFv) antibody developed to selectively bind pathological tau variants associated with AD and other tauopathies. Isolated via atomic force microscopy (AFM)-based biopanning, ADT-4 distinguishes AD biomarkers in human tissue and biofluids .

Key Research Findings:

  • Specificity: ADT-4 selectively binds tau aggregates in AD brain tissue but not in cognitively normal samples .

  • Diagnostic Utility:

    • In longitudinal plasma studies of 50 patients, ADT-4 detected elevated tau levels in ApoE3/3 AD cases compared to ApoE3/4 carriers .

    • Immunohistochemical analysis showed partial overlap with phosphorylated tau (AT8 antibody) in AD brain slices .

  • Therapeutic Potential:

    • ADT-4 reduced neuronal toxicity induced by AD-derived tau in SH-SY5Y cells, as measured by lactate dehydrogenase (LDH) assays .

Table 1: ADT-4 Performance in Clinical Samples

Study ParameterADT-4 OutcomeSource
Sensitivity (AD vs. control)100% discrimination in plasma samples
ApoE3/3 vs. ApoE3/4Higher tau levels in ApoE3/3 AD cases
Toxicity Reduction~40% protection in neuronal cell assays

ADT4 Antibody in Plant Biochemistry

ADT4 (Arogenate Dehydratase 4) is a chloroplastic enzyme antibody targeting phenylalanine biosynthesis in Arabidopsis thaliana. It is one of six ADT isoforms (ADT1–ADT6) identified in the Arabidopsis genome .

Comparative Analysis of ADT-4 and ADT4

While both antibodies share the "ADT4" designation, their targets and applications are distinct:

Table 3: ADT-4 vs. ADT4 Antibody Comparison

FeatureADT-4 (Tau scFv)ADT4 (Plant Enzyme)
TargetPathological tau aggregatesArogenate dehydratase 4 (ADT4)
SpeciesHumanArabidopsis thaliana
ApplicationNeurodegenerative disease diagnosticsPlant metabolic studies
Key StudyLongitudinal plasma analysis (n=50)Arabidopsis mutant phenotyping
Commercial SourceResearch-grade (non-commercial)PhytoAB (Catalog: PHY7541S)

Research Challenges and Future Directions

  • ADT-4 Limitations:

    • Partial overlap with phosphorylated tau suggests non-exclusive binding .

    • No protective effect observed in ADAMTS4 knock-out mouse models of osteoarthritis .

  • ADT4 Plant Studies:

    • Functional redundancy among six ADT isoforms complicates phenotype analysis .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
ADT4 antibody; At3g44720 antibody; T32N15.11Arogenate dehydratase 4 antibody; chloroplastic antibody; AtADT4 antibody; EC 4.2.1.91 antibody
Target Names
ADT4
Uniprot No.

Target Background

Function
This antibody targets an enzyme that catalyzes the conversion of prephenate (a product of the shikimate-chorismate pathway) to phenylalanine.
Database Links

KEGG: ath:AT3G44720

STRING: 3702.AT3G44720.1

UniGene: At.22683

Subcellular Location
Plastid, chloroplast stroma.
Tissue Specificity
Expressed in roots, leaves, stems, flowers and siliques. More abundant in stems and roots.

Q&A

What is ADAMTS4 and why is it a significant research target?

ADAMTS4, also known as aggrecanase-1 or ADMP-1, is an enzyme that cleaves aggrecan, a major cartilage proteoglycan, at the '392-Glu-|-Ala-393' site. This enzyme plays a crucial role in cartilage turnover and has been implicated in the pathogenesis of arthritic diseases through excessive cartilage breakdown . Additionally, ADAMTS4 may exacerbate neurodegeneration in Alzheimer's disease, broadening its significance as a research target .

The enzyme's balanced activity is essential for proper cartilage function and inflammation resolution in tissues, making it a compelling target for both basic research into tissue homeostasis and applied research into potential therapeutic interventions for inflammatory and degenerative conditions .

What applications are ADAMTS4 antibodies suitable for in research settings?

ADAMTS4 antibodies have demonstrated utility in multiple experimental applications, with validated protocols for:

  • Western Blot (WB): Using dilutions of approximately 1/3000 for detecting ADAMTS4 in cell lysates such as SH-SY5Y cells

  • Immunocytochemistry/Immunofluorescence (ICC/IF): Effective at 1/100 dilution for visualizing ADAMTS4 distribution in cellular contexts, such as in C6 cells with DAPI counterstaining for nuclei

  • Immunohistochemistry: For detecting ADAMTS4 in tissue sections, particularly in cartilage and neurological tissues where the protein is physiologically relevant

Most commercially available ADAMTS4 antibodies have been validated for mouse, rat, and human samples, facilitating comparative studies across species . When selecting an antibody for a specific application, researchers should verify the validation status for their particular experimental system.

How do I determine the optimal concentration of antibodies for ADAMTS4 detection?

Determining optimal antibody concentration is critical for balancing specific signal detection with minimal background. Research on antibody titration shows that:

  • Many antibodies reach signal saturation between 0.62 and 2.5 μg/mL, with higher concentrations primarily increasing background without improving specific detection

  • Antibodies used at concentrations at or below 0.62 μg/mL typically show a near-linear response to dilution, making them easier to optimize

  • Antibodies used at concentrations at or above 2.5 μg/mL often show minimal response to fourfold titration, suggesting diminishing returns at higher concentrations

For ADAMTS4 specifically, reported effective dilutions include 1/100 for immunofluorescence applications and 1/3000 for Western blotting . A systematic titration approach is recommended, starting with manufacturer-suggested dilutions and then testing 2-3 dilutions in both directions to identify the optimal signal-to-noise ratio for your specific experimental system.

How can ADAMTS4 antibodies be applied in studies of arthritic disease progression?

ADAMTS4 antibodies enable sophisticated investigations into arthritic disease mechanisms through multiple approaches:

  • Temporal expression analysis: Track ADAMTS4 levels throughout disease progression using quantitative immunohistochemistry or Western blotting with standardized loading controls

  • Co-localization studies: Combine ADAMTS4 antibodies with markers of inflammation or cartilage degradation to establish spatial and functional relationships

  • Therapeutic intervention assessment: Monitor changes in ADAMTS4 expression following treatments that target inflammatory pathways

  • Aggrecanase activity correlation: Pair ADAMTS4 protein detection with functional assays measuring aggrecan degradation to establish causative relationships

Since ADAMTS4 contributes significantly to disease progression through aggrecan degradation, antibody-based detection can help establish whether cartilage breakdown correlates with enzyme upregulation or post-translational activation . This information is valuable for determining optimal intervention points in the disease process.

What considerations are important when designing experiments to evaluate anti-ADAMTS4 therapeutic antibodies?

Developing therapeutic antibodies against ADAMTS4 requires careful experimental design addressing several key factors:

  • Epitope selection: Target regions that directly influence enzymatic activity rather than just binding to the protein

  • Specificity verification: Ensure the antibody discriminates between ADAMTS4 and related family members (particularly ADAMTS5, which has overlapping functions)

  • Neutralization assessment: Implement functional assays measuring aggrecan cleavage to confirm inhibition of enzymatic activity beyond simple binding

  • Immunogenicity evaluation: Design experiments to detect anti-drug antibody (ADA) formation, as ADAs can affect pharmacokinetics, safety, and efficacy

  • Pharmacokinetic/pharmacodynamic (PK/PD) modeling: Incorporate studies that relate antibody concentration to enzyme inhibition over time

How can researchers distinguish between the roles of ADAMTS4 and other aggrecanases in cartilage degradation models?

Differentiating the specific contributions of ADAMTS4 from other aggrecanases (particularly ADAMTS5) requires multifaceted experimental approaches:

  • Selective antibodies: Utilize antibodies validated for specificity against particular epitopes unique to ADAMTS4

  • Neoepitope antibodies: Employ antibodies that recognize the specific cleavage patterns in aggrecan produced by ADAMTS4 versus other aggrecanases

  • Gene silencing validation: Complement antibody studies with selective knockdown of ADAMTS4 using siRNA or CRISPR-Cas9 techniques

  • Knockout models: Compare findings from ADAMTS4-deficient models with wild-type controls to confirm antibody-based observations

  • Inhibitor specificity controls: Include experimental conditions with selective chemical inhibitors alongside antibody treatments

What approaches should be used to develop and optimize immunoassays for ADAMTS4 detection?

Developing robust ADAMTS4 immunoassays benefits significantly from systematic optimization approaches rather than traditional sequential testing:

  • Design of Experiments (DOE): Implement multivariate experimental design rather than univariate or bivariate experiments to efficiently optimize critical parameters including:

    • Concentrations of capture and detection reagents

    • Assay incubation times

    • Buffer compositions and blocking agents

  • Validation criteria: Establish comprehensive performance metrics for:

    • Sensitivity (limit of detection)

    • Specificity (cross-reactivity with related proteins)

    • Precision (intra- and inter-assay variability)

    • Accuracy (recovery of known concentrations)

    • Drug tolerance (ability to detect ADAMTS4 in the presence of potential interfering factors)

DOE approaches have demonstrated superior efficiency in immunoassay development compared to conventional checkerboard optimization, enabling simultaneous optimization of multiple parameters while accounting for potential interactions between variables .

What strategies minimize background signal when using ADAMTS4 antibodies in multimodal analyses?

Controlling background signal is particularly important for obtaining meaningful results in complex applications such as multimodal single-cell analysis:

  • Concentration optimization: Evidence shows that antibody concentrations above 2.5 μg/mL often contribute primarily to background rather than specific signal enhancement

  • Titration response monitoring: Antibodies used at concentrations below 0.62 μg/mL typically show linear response to dilution, making signal-to-noise optimization more predictable

  • Blocking protocol refinement: For Western blot applications, 3% nonfat dry milk in TBST has been effective for ADAMTS4 antibody applications

  • Signal quantification methods: For oligo-conjugated antibodies, monitor UMI (Unique Molecular Identifier) counts at the 90th quantile of the cell type with highest expression rather than total counts to better represent signal in positive populations

Quantitative data show that when antibody concentration was reduced fourfold (75% reduction), the corresponding decrease in UMI counts was only 38-51%, demonstrating diminishing returns at higher concentrations . This suggests starting with lower concentrations and gradually increasing as needed rather than defaulting to high concentrations.

How should researchers characterize antibody-antigen binding kinetics for ADAMTS4 studies?

Comprehensive characterization of antibody-antigen binding kinetics provides critical insights into antibody performance and potential therapeutic applications:

  • Key parameters to measure:

    • Association rate constant (kon)

    • Dissociation rate constant (koff)

    • Equilibrium dissociation constant (KD)

    • Binding stoichiometry

    • Temperature and pH dependence of binding

  • Recommended technologies:

    • Surface Plasmon Resonance (SPR) for real-time kinetic measurements

    • Bio-Layer Interferometry (BLI) for label-free analysis

    • Isothermal Titration Calorimetry (ITC) for thermodynamic profiling

    • Emerging computational methods for modeling physiologically relevant conditions

Understanding these kinetic parameters is crucial for predicting antibody performance in both research and therapeutic contexts, as binding kinetics represent key determinants of biological function and potential success as biotherapeutics . These characterizations provide essential foundation for later pharmacokinetic/pharmacodynamic (PK/PD) studies if pursuing therapeutic development .

How can researchers address inconsistent Western blot results with ADAMTS4 antibodies?

When facing inconsistent Western blot results for ADAMTS4 detection, systematic troubleshooting should address:

  • Sample preparation optimization:

    • Evaluate different lysis buffer compositions

    • Test various protease inhibitor cocktails to prevent degradation

    • Optimize protein loading (typically 20-50 μg total protein)

    • Consider non-reducing versus reducing conditions as epitope accessibility may differ

  • Detection protocol refinement:

    • Verify primary antibody dilution (1/3000 has been effective for ADAMTS4)

    • Optimize blocking conditions (3% nonfat dry milk in TBST is recommended)

    • Adjust incubation times and temperatures

    • Test alternative membrane types (PVDF vs. nitrocellulose)

  • Signal interpretation considerations:

    • ADAMTS4 may present as multiple bands due to proteolytic processing

    • Full-length ADAMTS4 (~90 kDa) versus processed forms

    • Post-translational modifications may affect migration patterns

    • Validate with positive control lysates (e.g., SH-SY5Y cells)

Maintaining detailed records of experimental conditions facilitates systematic optimization and ensures reproducibility once optimal conditions are established.

What factors might explain differences between ADAMTS4 levels detected by antibody-based methods versus functional activity assays?

Discrepancies between detected protein levels and measured enzymatic activity may arise from several factors:

  • Post-translational regulation mechanisms:

    • ADAMTS4 requires proteolytic activation to remove its pro-domain

    • Activity is modulated by glycosylation patterns

    • Phosphorylation state may affect catalytic efficiency without changing protein levels

  • Endogenous inhibitors:

    • Tissue inhibitors of metalloproteinases (TIMPs) may mask activity without affecting antibody detection

    • The balance between ADAMTS4 and its inhibitors is critical for proper cartilage function

  • Methodological considerations:

    • Antibodies may detect both active and inactive forms

    • Sample processing may activate latent enzyme or inactivate active enzyme

    • Assay conditions (pH, ionic strength, presence of cofactors) may not reflect in vivo activity

Addressing these discrepancies requires complementary approaches combining quantitative protein detection with activity-based assays under standardized conditions, and potentially direct analysis of aggrecan cleavage products.

How should researchers interpret data from anti-drug antibody (ADA) formation in ADAMTS4-targeting therapeutic studies?

When evaluating ADA formation in the context of ADAMTS4-targeting therapeutics, researchers should consider:

  • Differential impact assessment:

    • In clinical trials, only 27% of detected ADA formation was associated with altered pharmacodynamics or reduced efficacy

    • Some patients with high ADA titers still show therapeutic response, potentially due to low antibody affinity or timing of ADA formation

  • Methodological standardization:

    • Use consistent detection and reporting methods across studies

    • Implement sensitive assays that can detect clinically relevant ADA levels

    • Design of Experiments (DOE) approaches improve assay development efficiency

  • Interpretation framework:

    • Correlate ADA titers with pharmacokinetic parameters

    • Assess neutralizing versus non-neutralizing antibodies

    • Consider timing of ADA formation relative to treatment course

    • Evaluate epitope specificity of the ADAs

These considerations are essential because while 21% of trials found that ADAs had no effect on efficacy, the majority (51%) did not thoroughly explore these potential relationships , highlighting the importance of systematic ADA characterization in therapeutic antibody development.

What emerging technologies might enhance ADAMTS4 antibody applications in precision medicine?

Several cutting-edge technologies are poised to revolutionize ADAMTS4 antibody applications:

  • Advanced antibody engineering:

    • Bispecific antibodies targeting ADAMTS4 and complementary disease mediators

    • Site-specific antibody conjugation for more homogeneous antibody-drug conjugates (ADCs)

    • Computationally designed antibodies with optimized binding properties

  • Novel detection platforms:

    • Multimodal single-cell analysis combining protein and RNA detection

    • In situ protein detection with spatial resolution

    • High-throughput automated image analysis for tissue distribution studies

  • Therapeutic applications:

    • Antibody-drug conjugates (ADCs) delivering cytotoxic payloads to cells overexpressing ADAMTS4

    • Engineered antibody fragments with enhanced tissue penetration

    • Combination therapies targeting multiple aspects of disease pathways

These technologies will enable more precise targeting of ADAMTS4 in specific disease contexts and cell populations, potentially improving both research applications and therapeutic interventions.

How might computational approaches advance ADAMTS4 antibody development and application?

Computational methods represent a growing frontier in antibody research with several promising applications:

  • Epitope mapping and antibody design:

    • In silico prediction of optimal binding epitopes on ADAMTS4

    • Structure-based antibody design targeting functional domains

    • Molecular dynamics simulations to predict binding stability

  • Pharmacokinetic/pharmacodynamic modeling:

    • Prediction of antibody distribution and target engagement in different tissues

    • Modeling of dose-response relationships for therapeutic planning

    • Integration of binding kinetics data to predict in vivo efficacy

  • Data integration and analysis:

    • Machine learning approaches to correlate antibody properties with functional outcomes

    • Network analysis to understand ADAMTS4 in broader biological contexts

    • Predictive modeling of potential off-target effects

Emerging computational methods are becoming powerful tools for modeling antibody-binding interactions under physiologically relevant conditions, offering insights that may be difficult to obtain through experimental approaches alone .

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.