AGD2 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of ADG2 Antibody

ADG2 is a monoclonal antibody (mAb) developed for broad-spectrum protection against sarbecoviruses, including SARS-CoV and SARS-CoV-2. It exhibits pan-coronavirus neutralization capabilities, targeting conserved epitopes across viral variants ( ).

Mechanistic Insights

ADG2 functions by binding to a highly conserved epitope on the viral RBD, preventing receptor interaction. Structural studies confirm its ability to maintain binding despite mutations in circulating SARS-CoV-2 variants ( ).

Neutralization Performance

  • SARS-CoV-2 Variants: Neutralized >30 strains, including those resistant to other mAbs (e.g., Beta, Gamma).

  • Cross-Reactivity: Effective against bat coronaviruses WIV-1 and SHC014.

  • Potency: Comparable or superior to leading COVID-19 antibodies in development (e.g., REGN10987, LY-CoV555) ( ).

Preclinical Efficacy

In murine models, ADG2 demonstrated:

  • Prophylactic Protection: Complete protection against severe SARS-CoV-2 and SARS-CoV.

  • Therapeutic Potential: Reduced viral load in established infections.

Comparative Neutralization Data

Assay MetricADG2 Performance vs. Other mAbs
Half-maximal inhibitory concentration (IC₅₀)2–10× lower (indicating higher potency)
Epitope ConservationNo mutations reported in key contact residues (as of December 2020)

Advantages Over Existing Antibodies

  • Resistance Mitigation: Targets a conserved epitope, minimizing escape mutations.

  • Broad Applicability: Potential utility against future SARS-related viruses.

  • Manufacturability: Engineered for stability and scalable production ( ).

Research and Development Challenges

  • Immunogenicity Risk: Requires further evaluation in human trials.

  • Dosage Optimization: Balancing efficacy with potential Fc-mediated effects (e.g., antibody-dependent enhancement [ADE]) ( ).

Future Directions

ADG2’s design principles are being leveraged for pan-sarbecovirus vaccine development, aiming to elicit similar broadly neutralizing antibodies. Collaboration with structural biologists and AI-driven protein engineering (e.g., language models for antibody evolution) may enhance its clinical profile ( ).

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
AGD2 antibody; At1g60680 antibody; F8A5.20Probable aldo-keto reductase 2 antibody; EC 1.1.1.- antibody; ARF-GAP domain-containing protein 2 antibody
Target Names
AGD2
Uniprot No.

Q&A

What is AGD2 and why are antibodies against it important for research?

AGD2 (ARF-GAP DOMAIN 2) is a member of the ARF GAP domain family in Arabidopsis thaliana. It belongs to class 1 of AGD proteins, along with AGD1, AGD3, and AGD4 . AGD2 is also known as VAL2 (VAN3-LIKE PROTEIN2) and functions as a GTPase-activating protein. Antibodies against AGD2 are valuable research tools for studying ARF-GTPase signaling pathways in plants, particularly for investigating membrane trafficking, cell expansion, and development processes.

The importance of AGD2 antibodies stems from the need to:

  • Detect and quantify native AGD2 protein expression in different tissues

  • Analyze subcellular localization using immunohistochemistry techniques

  • Validate protein-protein interactions through co-immunoprecipitation

  • Monitor AGD2 expression changes during developmental stages or stress responses

What are the characteristics of commercially available AGD2 antibodies?

Commercial AGD2 antibodies typically exhibit these characteristics:

ParameterSpecificationsApplication Notes
Host SpeciesRabbit (polyclonal)Suitable for Western blot applications
ImmunogenKLH-conjugated synthetic peptide (16 aa from central section)Derived from Arabidopsis thaliana AGD2 (AT1G60860)
FormLyophilizedRequires reconstitution with 150 μl sterile water
Quantity~150 μg per vialSufficient for multiple experiments
Stability12 months at -20 to -70°C (as supplied)6 months after reconstitution when stored properly
Recommended Dilution1:1000-1:2000For Western blot applications
Expected MW88 kDaMay vary based on post-translational modifications

For optimal results, antibodies should be reconstituted according to manufacturer recommendations, with brief centrifugation prior to opening to prevent loss of lyophilized material .

How should AGD2 antibodies be stored and handled to maintain reactivity?

Proper storage and handling are critical for maintaining AGD2 antibody performance:

  • Before reconstitution:

    • Store lyophilized antibody at -20°C to -70°C

    • Avoid exposure to repeated temperature fluctuations

    • Protect from light and moisture

  • After reconstitution:

    • Store at -20°C to -70°C for long-term (up to 6 months)

    • For short-term use (up to 1 month), store at 2-8°C

    • Avoid repeated freeze-thaw cycles by preparing working aliquots

    • Centrifuge vials briefly before opening to collect all material

  • During experiments:

    • Keep on ice when in use

    • Return to appropriate storage immediately after use

    • Use sterile techniques to prevent contamination

Implementing these practices extends antibody shelf-life and ensures consistent experimental results.

What are the optimal Western blot conditions for detecting AGD2 protein?

Optimized Western blot protocol for AGD2 detection:

  • Sample preparation:

    • Extract total protein from plant tissues using a buffer containing protease inhibitors

    • Quantify protein concentration using Bradford or BCA assay

    • Load 20-50 μg of total protein per lane

  • Electrophoresis conditions:

    • Use 12% SDS-PAGE for optimal separation

    • Include positive controls (recombinant AGD2 protein at 2.5, 10, and 25 ng)

    • Include molecular weight markers that span 50-100 kDa range

  • Transfer parameters:

    • Transfer to nitrocellulose membrane at 100V for 1 hour

    • Verify transfer efficiency with reversible protein stain

  • Antibody incubation:

    • Block with 5% skim milk for 1 hour at room temperature

    • Incubate with primary AGD2 antibody at 1:1000 dilution overnight at 4°C

    • Wash 3× with TBST (10 minutes each)

    • Incubate with HRP-conjugated secondary antibody (anti-rabbit IgG) at 1:10,000 dilution for 1 hour at room temperature

    • Perform final washes (3× with TBST, 10 minutes each)

  • Detection:

    • Use enhanced chemiluminescence substrate

    • Optimize exposure time based on signal intensity (typically 30 seconds to 5 minutes)

    • Expected band at approximately 88 kDa

For troubleshooting weak signals, consider longer primary antibody incubation or higher antibody concentration (up to 1:500).

How can specificity of AGD2 antibodies be validated in experimental systems?

Rigorous validation of AGD2 antibody specificity is essential for reliable experimental results:

  • Peptide competition assays:

    • Pre-incubate AGD2 antibody with excess immunizing peptide (10-100× molar ratio)

    • Run parallel Western blots with blocked and unblocked antibody

    • Specific signals should disappear in the blocked antibody lane

  • Genetic validation approaches:

    • Test antibody against wild-type and agd2 knockout/knockdown plant tissues

    • Specific signals should be reduced or absent in knockout samples

    • Use CRISPR-edited lines with epitope modifications as additional controls

  • Cross-reactivity assessment:

    • Test against recombinant proteins from the related AGD family (AGD1, AGD3, AGD4)

    • Evaluate binding to proteins from other plant species to determine conservation

    • Perform epitope mapping to identify specific binding regions

  • Immunoprecipitation validation:

    • Perform IP followed by mass spectrometry to confirm target identity

    • Cross-validate with orthogonal detection methods (e.g., GFP-tagged AGD2)

What considerations are important when designing immunofluorescence experiments with AGD2 antibodies?

When planning immunofluorescence experiments using AGD2 antibodies, researchers should consider:

  • Fixation optimization:

    • Test multiple fixatives (4% paraformaldehyde, glutaraldehyde, methanol)

    • Optimize fixation time (15-30 minutes) and temperature

    • Evaluate epitope preservation through parallel Western blot analysis

  • Permeabilization parameters:

    • Optimize detergent type and concentration (0.1-0.5% Triton X-100 or 0.05-0.1% saponin)

    • Adjust permeabilization time based on tissue type (5-30 minutes)

    • Consider enzymatic cell wall digestion for plant tissues

  • Antibody incubation conditions:

    • Test concentration gradients (1:100 to 1:500)

    • Evaluate blocking solutions (BSA, normal serum, commercial blockers)

    • Determine optimal incubation time and temperature (overnight at 4°C vs. 2 hours at room temperature)

  • Controls and counterstaining:

    • Include no-primary antibody controls

    • Use known subcellular markers as co-staining references

    • Apply DAPI or other nuclear counterstains for orientation

  • Signal amplification options:

    • Consider tyramide signal amplification for low-abundance targets

    • Evaluate secondary antibody options (direct conjugates vs. biotin-streptavidin systems)

    • Test super-resolution microscopy techniques for detailed localization

Creating a systematic optimization matrix that varies these parameters will help establish reliable protocols for AGD2 visualization in different experimental contexts.

How should quantitative data from AGD2 antibody experiments be analyzed?

Rigorous quantitative analysis of AGD2 antibody experiments requires:

  • Western blot quantification:

    • Use technical replicates (minimum n=3) and biological replicates (minimum n=3)

    • Normalize AGD2 signal to appropriate loading controls (GAPDH, actin, tubulin)

    • Apply densitometry software with linear dynamic range verification

    • Create standard curves using recombinant AGD2 protein (2.5-25 ng range)

    • Report relative changes rather than absolute values when appropriate

  • Statistical analysis approaches:

    • Apply appropriate statistical tests based on data distribution (parametric vs. non-parametric)

    • Use ANOVA with post-hoc tests for multi-group comparisons

    • Report p-values and confidence intervals

    • Consider power analysis to determine adequate sample sizes

  • Visualization methods:

    • Present normalized data with error bars (standard deviation or standard error)

    • Include representative blot images alongside quantification

    • Use consistent scaling and cropping practices

    • Indicate molecular weight markers

  • Meta-analysis considerations:

    • Compare results across different experimental conditions

    • Evaluate consistency with published literature

    • Consider batch effects and antibody lot variations

These approaches align with modern antibody research practices that emphasize reproducibility and statistical rigor in data analysis .

How can contradictory results between different anti-AGD2 antibodies be reconciled?

When facing contradictory results between different AGD2 antibodies:

  • Epitope mapping analysis:

    • Determine the specific binding regions of each antibody

    • Analyze whether epitopes might be differentially accessible in various experimental conditions

    • Consider post-translational modifications that might affect epitope recognition

  • Antibody characterization comparison:

    • Review antibody production methods (monoclonal vs. polyclonal, host species)

    • Compare validation data from manufacturers

    • Evaluate specificity through peptide competition and knockout validation

  • Experimental variable assessment:

    • Systematically test buffer compositions, pH ranges, and detergent concentrations

    • Evaluate protein extraction methods (native vs. denaturing conditions)

    • Consider protein complex formation that might mask epitopes

  • Orthogonal validation approaches:

    • Implement non-antibody detection methods (mass spectrometry, RNA expression)

    • Use genetic approaches (tagged AGD2 constructs, CRISPR editing)

    • Apply proximity labeling techniques to confirm localization

  • Data integration framework:

    • Develop scoring systems that weight results based on validation quality

    • Implement Bayesian statistical approaches to incorporate prior knowledge

    • Consider computational modeling of antibody-epitope interactions

This systematic reconciliation approach helps distinguish between technical artifacts and genuine biological phenomena when interpreting contradictory results.

What standards should be applied when evaluating AGD2 antibody specificity claims?

When evaluating specificity claims for AGD2 antibodies, researchers should apply these standards:

Validation ParameterMinimum StandardGold Standard
Western BlotSingle band at expected MW (88 kDa)Band absent in knockout/knockdown samples
Peptide Competition>80% signal reductionComplete signal elimination
Cross-reactivityTested against AGD1, AGD3, AGD4Tested against full AGD family and species variants
ImmunoprecipitationTarget verification by mass specQuantitative enrichment analysis
Lot-to-Lot ConsistencyComparative WB analysisEpitope binding affinity measurements
Application ValidationTested in one applicationValidated across multiple applications

Critical evaluation factors should include:

  • Completeness of validation data provided by manufacturers

  • Concentration of antibody used in validation studies

  • Controls included in validation experiments

  • Whether validation was performed in the same experimental system as intended use

  • Publication record of the antibody in peer-reviewed literature

Researchers should document these evaluations to support reproducibility in antibody-based research .

How can AGD2 antibodies be modified for multiplex imaging applications?

Advanced multiplex imaging with AGD2 antibodies can be achieved through:

  • Direct labeling strategies:

    • Conjugate purified AGD2 antibodies with different fluorophores (Alexa Fluor 488, 555, 647)

    • Use NHS-ester chemistry for controlled dye-to-antibody ratios (optimal 4-6 dyes per antibody)

    • Implement zenon labeling for rapid, small-scale antibody labeling

    • Consider quantum dots for improved photostability in long-term imaging

  • Sequential staining approaches:

    • Apply tyramide signal amplification with spectrally distinct fluorophores

    • Implement iterative antibody stripping and restaining protocols

    • Use DNA-barcoded antibodies for highly multiplexed detection

  • Specialized multiplexing technologies:

    • Implement mass cytometry (CyTOF) using metal-conjugated AGD2 antibodies

    • Apply spectral unmixing algorithms for closely spaced fluorophores

    • Consider CODEX or similar spatial proteomics approaches

  • Dual-modality imaging strategies:

    • Combine fluorescence with electron microscopy using nanogold-conjugated antibodies

    • Implement correlative light and electron microscopy workflows

    • Develop PET/fluorescence dual-labeled antibodies similar to approaches used for other targets

  • Analysis considerations:

    • Apply automated image segmentation algorithms

    • Implement machine learning for pattern recognition

    • Use spatial statistics to quantify co-localization

These advanced approaches enable simultaneous visualization of AGD2 with interacting proteins, providing deeper insights into functional relationships in complex cellular processes.

What are the considerations for developing engineered anti-AGD2 antibodies with customized specificities?

Developing engineered AGD2 antibodies with custom specificities requires:

  • Epitope selection strategies:

    • Perform computational analysis of AGD2 sequence to identify unique regions

    • Select regions with high antigenicity and surface accessibility

    • Consider evolutionary conservation for cross-species reactivity

    • Target regions that distinguish AGD2 from other AGD family members

  • Antibody engineering approaches:

    • Implement CDR grafting techniques similar to those used for humanization

    • Apply phage display selection with specific binding/elution conditions

    • Use deep mutational scanning to identify specificity-enhancing mutations

    • Implement affinity maturation through directed evolution

  • Specificity optimization:

    • Conduct negative selection against related AGD proteins

    • Implement computational models for specificity prediction

    • Apply energy functions to model binding interactions

    • Design antibodies that recognize specific post-translational modifications

  • Validation pipeline:

    • Test against panels of recombinant AGD proteins

    • Measure binding kinetics using surface plasmon resonance

    • Perform epitope binning to confirm target regions

    • Validate in multiple experimental systems

  • Production considerations:

    • Optimize expression systems for consistent yields

    • Implement quality control metrics for batch consistency

    • Consider stability engineering for improved shelf-life

These approaches draw from established antibody engineering principles while addressing the specific challenges of developing highly selective AGD2 antibodies .

How can high-throughput screening approaches be applied to AGD2 antibody characterization?

Implementing high-throughput screening for AGD2 antibody characterization:

  • Next-generation sequencing integration:

    • Apply NGS to characterize antibody repertoires from immunized animals

    • Sequence CDR regions to identify diversity in binding domains

    • Use bioinformatic clustering to identify antibody families

    • Implement sequence-function correlations to predict binding properties

  • Microarray-based epitope mapping:

    • Create peptide arrays covering the complete AGD2 sequence

    • Test antibody binding across overlapping peptides

    • Identify critical binding residues through alanine scanning

    • Create heat maps of binding intensity across AGD2 sequence regions

  • Automated screening platforms:

    • Implement liquid handling systems for antibody dilution series

    • Use automated Western blot systems for consistency

    • Apply robotic microscopy for immunofluorescence screening

    • Develop high-content analysis workflows for quantitative assessment

  • Design of experiments approaches:

    • Apply DOE principles to systematically vary experimental parameters

    • Create response surface models to identify optimal conditions

    • Use factorial designs to assess interaction effects

    • Implement quality by design principles from antibody development

  • Computational analysis integration:

    • Apply machine learning for pattern recognition in binding data

    • Develop predictive models for antibody performance

    • Implement cluster analysis to identify antibody groups with similar properties

    • Create visualization tools for complex dataset interpretation

These high-throughput approaches accelerate the characterization process while providing more comprehensive data on AGD2 antibody performance characteristics across multiple parameters simultaneously.

How might single-cell analysis techniques be integrated with AGD2 antibody applications?

Integrating AGD2 antibodies with single-cell analysis enables more nuanced understanding of cellular heterogeneity:

  • Single-cell Western blot applications:

    • Apply microfluidic platforms for single-cell protein analysis

    • Quantify AGD2 expression variability within populations

    • Correlate with cell morphological parameters

    • Develop miniaturized Western blot protocols optimized for AGD2 detection

  • Mass cytometry integration:

    • Conjugate AGD2 antibodies with rare earth metals

    • Combine with markers for cell type, cell cycle, and signaling pathways

    • Implement computational algorithms for high-dimensional data analysis

    • Develop clustering approaches to identify cell subpopulations

  • Spatial transcriptomics combination:

    • Correlate AGD2 protein localization with gene expression patterns

    • Implement sequential IF and in situ hybridization protocols

    • Develop computational frameworks for multi-omic data integration

    • Apply tissue clearing techniques for whole-organ analysis

  • Single-cell proteomics approaches:

    • Validate AGD2 antibodies for nanobody-based proximity labeling

    • Implement scProteomics workflows incorporating AGD2 detection

    • Develop calibration standards for absolute quantification

    • Apply trajectory analysis for temporal protein dynamics

These integrated approaches will provide unprecedented resolution in understanding AGD2 function across diverse cellular contexts and developmental stages.

What considerations are important when developing AGD2 antibodies for super-resolution microscopy?

Optimizing AGD2 antibodies for super-resolution microscopy applications:

  • Fluorophore selection criteria:

    • Choose fluorophores with high photon yields and appropriate photoswitching properties

    • Evaluate quantum yield and extinction coefficient parameters

    • Consider photobleaching rates for time-lapse applications

    • Test dye pairs for STORM/PALM applications

  • Conjugation optimization:

    • Control labeling density to achieve appropriate spatial resolution

    • Implement site-specific conjugation strategies to maintain binding properties

    • Develop small-format antibody derivatives (Fabs, nanobodies) for improved resolution

    • Optimize dye-to-antibody ratios for specific super-resolution techniques

  • Sample preparation considerations:

    • Develop specialized fixation protocols that preserve nanoscale structures

    • Optimize clearing protocols compatible with AGD2 epitopes

    • Implement expansion microscopy approaches for physical magnification

    • Test different mounting media for optimal photophysical properties

  • Validation approaches:

    • Confirm specificity at super-resolution level using knockout controls

    • Verify localization patterns with orthogonal techniques

    • Develop quantitative metrics for resolution and specificity

    • Implement correlative electron microscopy for structural validation

  • Analysis workflows:

    • Apply specialized clustering algorithms for quantitative analysis

    • Develop drift correction protocols for long acquisition times

    • Implement computational approaches for localization precision estimation

    • Create visualization tools appropriate for sub-diffraction data

These optimizations will enable researchers to study AGD2 localization and dynamics at nanoscale resolution, providing new insights into its functional organization.

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.