AFB3 Antibody

Shipped with Ice Packs
In Stock

Description

Western Blot Validation

The antibody has been validated for Western Blot using cell lysates, demonstrating high specificity for AFF3 . Its cross-reactivity spans multiple species, including:

  • Human: 100% predicted reactivity

  • Mouse: 93%

  • Rat: 93%

  • Dog: 86%

This broad reactivity facilitates comparative studies across models.

Antibody Performance

  • Sensitivity: Detects AFF3 at low concentrations in WB.

  • Specificity: Minimal cross-reactivity with non-target proteins due to affinity purification .

  • Buffer: Supplied in PBS with 0.09% sodium azide and 2% sucrose for stability .

Comparative Analysis of AFF3 Antibodies

The AFF3 antibody (ABIN2778345) is one of several clones available, differing in epitope targets and applications:

CloneEpitope RegionHostApplications
ABIN2778345Middle (AA 1197–1227)RabbitWB
RB1916C-TerminalRabbitWB, IHC
NI-1701CD47/CD19HumanImmune checkpoint studies

This antibody is preferred for WB due to its high specificity and middle-region targeting, reducing interference from post-translational modifications .

Limitations and Future Directions

  • Species Limitation: No reactivity reported in non-mammalian models.

  • Therapeutic Potential: While AFF3 antibodies are primarily research tools, bispecific antibodies (e.g., AFM13 targeting CD30/CD16A) highlight the potential for clinical translation in oncology .

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
AFB3 antibody; At1g12820 antibody; F13K23.7 antibody; Protein AUXIN SIGNALING F-BOX 3 antibody
Target Names
AFB3
Uniprot No.

Target Background

Function
This antibody confers sensitivity to the virulent bacterial pathogen *Pseudomonas syringae*. It is a component of SCF (SKP1-cullin-F-box) E3 ubiquitin ligase complexes, which may mediate the ubiquitination and subsequent proteasomal degradation of target proteins. As an auxin receptor, it mediates the proteasomal degradation of Aux/IAA proteins and auxin-regulated transcription. It is also involved in the regulation of embryogenesis by auxin.
Gene References Into Functions
  1. AFB3 is capable of activating two independent pathways to control root system architecture. PMID: 23847199
Database Links

KEGG: ath:AT1G12820

STRING: 3702.AT1G12820.1

UniGene: At.23978

Subcellular Location
Nucleus.
Tissue Specificity
Ubiquitous, with higher levels in flowers.

Q&A

What is APBB3 and why are antibodies against it important in research?

APBB3 (Amyloid-beta A4 precursor protein-binding family B member 3) is a human protein that plays roles in various cellular processes. Antibodies against APBB3, such as polyclonal rabbit antibodies, are important research tools for studying protein expression, localization, and function in human tissues and cell lines . These antibodies enable researchers to detect and quantify APBB3 across multiple experimental applications including immunohistochemistry (IHC), immunocytochemistry (ICC), and Western blotting (WB). The significance of APBB3 research stems from its potential involvement in cellular pathways relevant to neurodegenerative conditions and other diseases, making specific antibodies crucial tools for investigating its biological functions.

What validation methods should be employed before using a new APBB3 antibody?

Before incorporating a new APBB3 antibody into research protocols, comprehensive validation is essential to ensure specificity, sensitivity, and reproducibility. Rigorous validation methods include:

  • Western blot analysis: Confirm the antibody detects bands of expected molecular weight in relevant tissues or cell lines

  • Immunohistochemistry (IHC): Verify expected staining patterns in tissues known to express APBB3

  • Positive and negative controls: Use tissues or cell lines with known APBB3 expression profiles

  • Blocking peptide experiments: Ensure signal disappears when the antibody is pre-incubated with its target antigen

  • Cross-reactivity testing: Check for non-specific binding to related proteins

Advanced validation should include siRNA knockdown or CRISPR knockout experiments to confirm specificity in biological systems . Many antibody manufacturers now provide enhanced validation data, including specificity testing across different applications to ensure the antibody performs consistently across experimental conditions.

How do polyclonal and monoclonal antibodies differ in research applications?

CharacteristicPolyclonal AntibodiesMonoclonal Antibodies
SourceMultiple B-cell clonesSingle B-cell clone
Epitope recognitionMultiple epitopesSingle epitope
Batch-to-batch variationHigherLower
SensitivityOften higher due to multiple epitope bindingMay be lower but more specific
Production complexityLess complex, fasterMore complex, time-consuming
Research applicationsGood for proteins expressed at low levels; robust signal detectionExcellent for distinguishing between closely related proteins; consistent results
Impact of epitope changesLess affected by small changes in antigen structureMay lose binding with minor epitope alterations

When selecting between polyclonal antibodies (like the APBB3 antibodies in search results) and monoclonal antibodies, researchers should consider the experimental context . Polyclonal antibodies often provide stronger signals due to binding multiple epitopes, making them useful for detecting proteins with low expression. Monoclonal antibodies offer superior specificity and consistency across experiments but may be less sensitive or fail if their single epitope is masked or altered in the experimental system.

How is AlphaFold3 changing antibody design and research?

AlphaFold3 (AF3) represents a significant advancement in computational antibody modeling, offering researchers new capabilities that accelerate therapeutic antibody development. AF3 demonstrates notable improvements in two critical areas:

  • Antibody-antigen docking prediction: AF3 achieves 8.9% high-accuracy docking success for antibodies and 13.4% for nanobodies, outperforming previous computational methods .

  • Structure prediction accuracy: For the critical CDR H3 loop region (complementarity-determining region), AF3 achieves median RMSD accuracy of 2.04 Å for antibodies and 1.14 Å for nanobodies .

These capabilities are transforming antibody research by:

  • Accelerating lead candidate identification: Researchers can computationally screen antibody candidates before experimental validation

  • Improving rational design approaches: Better structure prediction enables more precise antibody engineering

  • Enhancing understanding of binding mechanisms: Accurate modeling reveals key interaction residues

Despite these advances, AF3 still has limitations, with approximately 60% failure rate in antibody docking when using a single seed . Researchers are developing complementary approaches like enhanced sampling techniques to address these limitations. For researchers without direct access to AlphaFold3, alternatives like the open-source Boltz-1 service offer similar functionality for antibody structure prediction and docking analysis .

What approaches exist for designing antibodies with custom specificity profiles?

Designing antibodies with tailored specificity profiles requires sophisticated experimental and computational approaches. Recent advances combine high-throughput selection methods with biophysics-informed computational modeling to create antibodies with precisely controlled binding properties .

The process typically involves:

  • Initial library generation and selection: Creating diverse antibody libraries and selecting candidates through phage display against target and non-target antigens

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

  • Energy function optimization: For cross-specific antibodies, jointly minimizing energy functions associated with desired ligands; for highly specific antibodies, minimizing energy for the target while maximizing energy for non-targets

  • Experimental validation: Testing computationally designed candidates through binding assays

One successful approach demonstrated in recent research utilized a minimal antibody library with systematic variation in the CDR3 region, followed by computational analysis to disentangle binding modes associated with chemically similar ligands . This method successfully generated antibodies with customized specificity profiles not present in the original experimental library.

The ability to computationally design antibodies with predefined binding profiles offers particular value when:

  • Discriminating between very similar epitopes

  • Developing diagnostics that require precise antigen discrimination

  • Creating therapeutic antibodies that must avoid off-target binding

  • Designing cross-reactive antibodies for targeting multiple variants of pathogens

What are the advantages and limitations of different antibody fragment formats in research?

Antibody fragments offer distinct advantages for specific research applications compared to whole IgG molecules. Understanding their properties is crucial for experimental design:

FormatStructureMolecular WeightKey AdvantagesLimitationsBest Applications
Whole IgGComplete antibody with Fc region and two Fab regions~160 kDaCost-effective, standard detection, longest half-lifeLarge size limits tissue penetration, Fc interactions may cause backgroundStandard immunodetection, long-term in vivo studies
F(ab')2Two antigen-binding fragments connected by disulfide bonds~110 kDaEliminates Fc-mediated effects, maintains bivalent bindingMore expensive than whole IgG, shorter half-lifeLive cell applications with Fc receptor-expressing cells
FabSingle antigen-binding fragment~50 kDaSmall size, good tissue penetration, monovalent bindingReduced avidity, shorter half-life, higher costApplications requiring tissue penetration, monovalent binding
VHH (Nanobody)Single-domain antibody fragment~15 kDaVery small size, excellent tissue penetration, stable in harsh conditionsLimited commercial availability, potential immunogenicityAccessing sterically restricted epitopes, intracellular targeting

Each format presents specific advantages for particular experimental contexts . For example, when working with live cells expressing Fc receptors, F(ab')2 fragments avoid unwanted binding to these receptors while maintaining bivalent antigen binding. VHH fragments (nanobodies) offer exceptional stability and the ability to access epitopes inaccessible to larger antibody formats, making them valuable for specialized applications .

The choice between formats should be guided by experimental requirements including the need for tissue penetration, presence of Fc receptors in the experimental system, and whether monovalent or bivalent binding is preferred.

How can researchers detect early and specific antibody responses in immunological studies?

Detecting early and specific antibody responses presents significant challenges in immunological research, particularly for infectious disease studies. Traditional methods often fail to capture the earliest antibody responses, as demonstrated in Lyme disease research where specific antibodies to outer surface proteins were historically considered undetectable until months after infection .

Advanced methodological approaches to detect early antibody responses include:

  • Immune complex dissociation techniques: These methods separate antigen-antibody complexes that may sequester antibodies, making them undetectable by conventional assays. In Lyme disease research, this approach revealed specific antibodies to OspA much earlier than previously thought possible .

  • Enhanced sensitivity Western blots: Techniques like biotin-avidin Western blots provide significantly higher sensitivity than conventional methods, enabling detection of low-abundance antibodies in early immune responses .

  • Recombinant antigen confirmation: Using recombinant versions of specific antigens (like recombinant OspA) confirms the specificity of detected antibodies and eliminates cross-reactivity concerns .

  • Isotype-specific analysis: Examining specific antibody isotypes (particularly IgM) can reveal early responses that might be missed when measuring total antibody levels .

These approaches have revealed that specific antibody responses may occur much earlier than previously detected, challenging established paradigms about the timing of humoral immunity. For example, in Lyme disease patients, specific antibodies to OspA were detected in patients with erythema migrans (the earliest sign of infection), contradicting the previous understanding that such antibodies appeared only after 6 months .

What strategies exist for improving the specificity of bispecific antibodies?

Bispecific antibodies (BsAbs) represent an advanced class of therapeutic molecules designed to simultaneously bind two different antigens. Improving their specificity requires sophisticated engineering approaches:

  • Heterodimer recombination platforms: Technologies like the ART-Ig platform promote proper chain pairing by introducing complementary charge differences in the Fc region. For example, one chain might incorporate (D360K, D403K) mutations while the other incorporates (K402D, K419D) mutations .

  • Common light chain strategies: Using identical light chains for both binding specificities reduces mispairing issues and improves manufacturing consistency .

  • Computational structure prediction: Advanced modeling tools like AlphaFold3 can predict structural compatibility and potential cross-reactivity issues before experimental testing .

  • CDR engineering: Precise modifications to complementarity-determining regions can fine-tune binding specificity while maintaining affinity.

  • Affinity balancing: Careful adjustment of binding affinities for each target can optimize therapeutic index and reduce off-target effects.

These approaches address the main challenges in bispecific antibody development, including chain mispairing, manufacturing heterogeneity, and unintended cross-reactivity. Recent examples like ERY947, which targets both GPC3 and CD3, demonstrate the potential of these technologies for creating highly specific therapeutic antibodies .

How should researchers optimize antibody validation across different experimental techniques?

Comprehensive antibody validation across multiple experimental techniques is essential for research reliability. An optimized validation strategy should include:

  • Application-specific validation: Each technique (IHC, ICC, WB, ELISA, etc.) places different demands on antibody performance. Validation should be performed separately for each intended application .

  • Orthogonal validation: Confirm target expression using independent methods such as mRNA detection (qPCR, RNA-seq) to corroborate protein detection results.

  • Genetic validation: Use genetic approaches (knockdown, knockout, overexpression) to confirm specificity:

    • siRNA knockdown: Reduces target expression

    • CRISPR knockout: Eliminates target expression

    • Overexpression: Increases target levels in systems with low endogenous expression

  • Physiologically relevant systems: Validate in experimental models that reflect the biological context of the research question.

  • Reproducibility testing: Verify consistency across:

    • Different lots of the same antibody

    • Different experimental protocols

    • Different laboratories

  • Titration experiments: Determine optimal antibody concentration for each application to maximize signal-to-noise ratio.

  • Cross-reactivity assessment: Test against related proteins or in systems known not to express the target.

Antibody manufacturers increasingly provide extensive validation data across applications, but researchers should independently verify performance in their specific experimental systems . Documentation of validation results is crucial for research reproducibility and should be included in publications and shared resources.

How might integrated computational-experimental approaches transform antibody engineering?

The integration of computational modeling with experimental selection represents a transformative approach to antibody engineering. This combined methodology offers several advantages over traditional approaches:

  • Expanded design space exploration: Computational methods can evaluate potential sequences beyond those accessible through experimental libraries alone, which are inherently limited in size and diversity .

  • Disentangling of binding modes: Advanced modeling can identify distinct binding mechanisms associated with specific targets, even when these targets are chemically similar and difficult to distinguish experimentally .

  • Custom specificity profile design: The ability to generate antibodies with precisely defined binding properties, either with high specificity for a single target or controlled cross-reactivity across multiple targets .

  • Reduction of experimental artifacts: Computational approaches can help mitigate biases and artifacts inherent in selection experiments .

  • Structure-guided optimization: Models like AlphaFold3 provide structural insights that guide rational engineering of binding interfaces .

The future trajectory of this field likely involves:

  • Multi-objective optimization: Simultaneously optimizing multiple antibody properties (affinity, specificity, stability, manufacturability)

  • Incorporation of dynamics: Moving beyond static structure prediction to model conformational flexibility

  • Machine learning from experimental data: Training models on large experimental datasets to improve predictive accuracy

  • Automated design-build-test cycles: Integrating computational design with automated experimental testing in rapid iteration cycles

These approaches have particular promise for addressing challenging targets like membrane proteins, transient conformational epitopes, and highly similar antigens that have historically been difficult to address with conventional methods .

What are the emerging approaches for enhancing antibody docking prediction accuracy?

Despite advances in computational antibody modeling, accurate prediction of antibody-antigen docking remains challenging, with even advanced models like AlphaFold3 showing significant failure rates. Emerging approaches to enhance docking prediction accuracy include:

These approaches are particularly important as research moves toward more challenging targets where proper docking prediction is essential for therapeutic development. Combining multiple complementary methods often yields the best results, suggesting that integrated prediction pipelines represent the most promising path forward .

How can researchers address common issues with antibody specificity and background signal?

Antibody specificity problems and excessive background signal represent common challenges in immunodetection experiments. Methodological solutions include:

  • For high background in immunohistochemistry/immunocytochemistry:

    • Optimize blocking conditions: Test different blocking agents (BSA, normal serum, commercial blockers)

    • Adjust antibody concentration: Perform titration experiments to find optimal dilution

    • Increase washing duration and stringency

    • Use specialized detergents appropriate for the application

    • Consider autofluorescence quenching reagents for fluorescent detection

  • For non-specific bands in Western blotting:

    • Increase membrane blocking time

    • Optimize antibody concentration

    • Increase washing stringency

    • Use gradient gels for better protein separation

    • Consider alternative antibody formats (e.g., F(ab')2 fragments to reduce Fc-mediated binding)

  • For cross-reactivity issues:

    • Pre-absorb antibody with related antigens

    • Use knockout/knockdown controls

    • Perform peptide competition assays

    • Consider alternative antibodies recognizing different epitopes

  • For inconsistent results between techniques:

    • Verify that the epitope is accessible in each application

    • Check for denaturation effects on epitope recognition

    • Ensure proper sample preparation for each technique

    • Validate the antibody specifically for each application

These methodological adjustments should be systematically tested and documented to establish reliable protocols. When selecting antibodies, researchers should prioritize those with validation data specifically relevant to their intended application and experimental system .

What considerations are important when designing experiments with bispecific antibodies?

Bispecific antibody (BsAb) experiments require special considerations beyond those for conventional antibodies. Key methodological aspects include:

  • Format selection: Choose the appropriate BsAb format (IgG-like, fragment-based, etc.) based on:

    • Size requirements and tissue penetration needs

    • Desired valency for each target

    • Required half-life and pharmacokinetics

    • Expression system compatibility

  • Control selection:

    • Include monospecific antibodies against each target

    • Consider "null" bispecific controls with one non-functional binding site

    • Use isotype-matched controls with irrelevant specificities

  • Binding assessment:

    • Verify binding to each target independently

    • Confirm simultaneous binding capability

    • Assess potential avidity effects

    • Measure binding kinetics and affinity for each target

  • Functional validation:

    • Test in relevant cell-based assays

    • Verify specific mechanisms (e.g., T-cell redirection, receptor crosslinking)

    • Assess potential Fc-mediated effects if the format includes an Fc region

  • Stability and manufacturing considerations:

    • Evaluate chain pairing efficiency and product homogeneity

    • Assess thermal and pH stability

    • Determine expression yields and purification approaches

    • Check for aggregation propensity

Advanced BsAb platforms like ART-Ig utilize charge-based mutations (e.g., D360K, D403K in one chain and K402D, K419D in the other) to promote correct chain association . These engineering approaches minimize mispairing issues that can compromise specificity and function. When designing experiments, researchers should carefully consider the specific mechanism of action intended for their BsAb and include appropriate controls to validate this mechanism.

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.