BEE3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
BEE3 antibody; BHLH50 antibody; EN76 antibody; At1g73830 antibody; F25P22.25Transcription factor BEE 3 antibody; Basic helix-loop-helix protein 50 antibody; AtbHLH50 antibody; bHLH 50 antibody; Protein Brassinosteroid enhanced expression 3 antibody; Transcription factor EN 76 antibody; bHLH transcription factor bHLH050 antibody
Target Names
BEE3
Uniprot No.

Target Background

Function
Positive regulator of brassinosteroid signaling.
Database Links

KEGG: ath:AT1G73830

STRING: 3702.AT1G73830.1

UniGene: At.34949

Subcellular Location
Nucleus.
Tissue Specificity
Expressed in stems.

Q&A

What is BR3 and why is it an important target for antibody development?

BR3 (B-cell activating factor receptor) is a specific receptor for BAFF (B-cell-activating factor belonging to the tumor necrosis factor family) expressed on all mature B cells. It plays a critical role in B-cell survival and maturation processes . The importance of BR3 as an antibody target stems from its involvement in B-cell-mediated diseases, making it valuable for both research applications and potential therapeutic development.

Methodologically, researchers targeting BR3 should consider:

  • The receptor's expression pattern across different B-cell developmental stages

  • Species-specific differences in BR3 structure (human and murine BR3 share only 52% identity in their extracellular domains)

  • The interaction between BR3 and its natural ligand BAFF, which can be mimicked or blocked by engineered antibodies

What are the primary research applications for anti-BR3 antibodies?

Anti-BR3 antibodies serve several critical research functions:

  • Cellular phenotyping: BR3 antibodies like BR3 and BR31 enable identification of B-lymphocyte populations in flow cytometry applications. BR3 staining patterns closely correlate with anti-human Ig staining in both peripheral blood and lymphoid tissues .

  • Functional studies: BR3 antibodies can be used to investigate B-cell survival, proliferation, and maturation pathways. Antagonistic BR3 antibodies can block BAFF-dependent human B-cell proliferation in vitro, enabling mechanistic studies of B-cell regulation .

  • In vivo modeling: BR3 antibodies effective at reducing murine B-cell populations in vivo provide tools for modeling B-cell depletion therapies and investigating B-cell roles in disease models .

  • Receptor-ligand interaction studies: Anti-BR3 antibodies that mimic BAFF binding (like CB3s) offer insights into receptor activation mechanisms .

How can researchers distinguish between specific and non-specific binding when using BR3 antibodies?

Ensuring specificity is fundamental to reliable antibody-based research. For BR3 antibodies, several methodological approaches help distinguish specific from non-specific binding:

  • Cell population analysis: Test antibodies against highly purified populations of B lymphocytes versus granulocytes, monocytes, and E+ lymphocytes. Antibodies like BR3 and BR31 demonstrate specificity by staining B lymphocytes while failing to react with other cell types .

  • Comparative staining patterns: Compare BR3 antibody staining with established B-cell markers like anti-human Ig. True BR3 staining should show strong correlation with these markers in both peripheral blood and lymphoid tissues .

  • Subpopulation identification: Selective staining of subpopulations (e.g., BR31 recognizes approximately 50% of B lymphocytes) can further validate specificity .

  • Cross-reactivity testing: Evaluate potential cross-reactivity with other TNF-family receptors to confirm target selectivity.

How can cross-species reactivity of BR3 antibodies be engineered and evaluated?

Cross-species reactive antibodies are valuable for translational research, enabling consistent targeting across model systems and humans. The development of cross-reactive BR3 antibodies illustrates key methodological approaches:

  • Affinity maturation strategy: Starting with an antibody showing preferential binding to one species (e.g., CB1 has μM affinity for murine BR3 but weak affinity for human BR3), researchers can employ affinity maturation to enhance binding to both species. CB3s, an affinity-matured variant of CB1, achieved sub-nM affinity for BR3 from both species .

  • Structural analysis approach: Alanine scanning and crystallographic structural analysis of antibody-receptor complexes (e.g., CB3s/BR3) reveal binding epitopes. Focus on conserved regions between species can guide engineering efforts .

  • Functional validation: Cross-species antibodies should be evaluated not only for binding but also for functional activity across species. For BR3 antibodies, this includes testing BAFF-blocking ability and B-cell modulation in both human and murine systems .

  • Phage library selection strategy: To identify cross-reactive antibodies, researchers can employ synthetic antibody phage libraries with alternating selection on human and murine targets .

What approaches can be used to develop bispecific antibodies involving BR3 targeting?

Bispecific antibodies (BsAbs) targeting BR3 along with a second target offer enhanced therapeutic potential. Development involves several sophisticated methodological approaches:

  • Heavy chain heterodimerization technologies:

    • ART-Ig platform: Introduces complementary charged mutations in the Fc region (e.g., D360K/D403K in one chain and K402D/K419D in the other) to promote heterodimer formation

    • SEED (Strand-Exchange Engineered Domain): Employs alternating segments from human IgA and IgG CH3 sequences to create heterodimers with predictable assembly

    • BEAT (Bispecific Engagement by Antibodies based on T-cell receptor): Mimics natural association of T-cell receptor α and β chains for selective pairing

  • Light chain mispairing prevention:

    • Orthogonal Fab interface: Introduces mutations creating complementary interfaces that enforce correct light chain pairing

    • Common light chain approach: Uses a single light chain that functions effectively with both heavy chains

  • Post-production processing:

    • Controlled Fab-arm exchange (cFAE): Exploits the natural process of Fab arm exchange seen in IgG4, adapted for IgG1 through specific mutations (F405L/K409R), allowing half-antibody exchange under reducing conditions

  • Functional validation: Testing bispecific constructs for:

    • Binding to both targets individually and simultaneously

    • Functional effects (e.g., BR3 pathway modulation)

    • Stability and manufacturability

How do antibody sequence diversity considerations impact BR3 antibody discovery and optimization?

Understanding antibody sequence diversity is crucial for efficient BR3 antibody discovery and optimization:

What are the optimal protocols for validating BR3 antibody specificity in flow cytometry?

Flow cytometry is a primary application for BR3 antibodies, requiring careful validation protocols:

  • Cell population comparison methodology:

    • Test against highly purified B lymphocytes (positive control)

    • Confirm lack of reactivity with granulocytes, monocytes, and E+ lymphocytes (negative controls)

    • Include comparative staining with established B-cell markers like anti-human Ig

  • Titration optimization:

    • Prepare logarithmic dilution series of the antibody

    • Identify concentration providing maximum specific signal with minimal background

    • Determine optimal dilution for each specific flow cytometer and fluorophore combination

  • Co-staining approach:

    • Combine BR3 antibody with antibodies against other B-cell markers

    • Analyze co-expression patterns to confirm expected cellular distribution

    • Verify staining patterns in both peripheral blood and lymphoid tissue samples

  • Blocking validation:

    • Pre-incubate with unlabeled antibody or recombinant BR3

    • Specific binding should be competitively inhibited

    • Analyze shift in staining pattern to confirm specificity

What techniques are most effective for characterizing BR3 antibody epitopes?

Detailed epitope characterization is essential for understanding BR3 antibody function:

  • Alanine scanning mutagenesis:

    • Systematically replace individual amino acids in BR3 with alanine

    • Test antibody binding to each mutant

    • Identify critical residues for the interaction

    • This approach helped characterize how CB3s mimics BAFF by interacting with a similar region of the BR3 surface

  • Crystallographic structural analysis:

    • Determine three-dimensional structure of antibody-BR3 complex

    • Identify specific contacts between antibody CDRs and BR3 epitopes

    • Compare binding mode to natural BAFF-BR3 interaction

    • This method provided structural insights into the CB3s/BR3 complex

  • Competition binding assays:

    • Test whether antibody competes with BAFF for BR3 binding

    • Determine if multiple antibodies can bind simultaneously or compete

    • Map relative epitope locations based on competition patterns

  • Hydrogen-deuterium exchange mass spectrometry:

    • Map regions of BR3 protected from solvent exchange upon antibody binding

    • Identify conformational changes in BR3 induced by antibody

    • Provide epitope information without requiring crystallization

What methodologies are recommended for evaluating BR3 antibody affinity?

Accurate affinity determination is critical for BR3 antibody characterization and optimization:

  • Surface plasmon resonance (SPR) protocol:

    • Immobilize purified BR3 or antibody on sensor chip

    • Measure real-time binding kinetics (kon and koff rates)

    • Calculate equilibrium dissociation constant (KD)

    • Compare affinity across species (human vs. murine BR3)

    • Evaluate affinity improvements in engineered variants (e.g., CB1 vs. CB3s)

  • Bio-layer interferometry (BLI) approach:

    • Alternative optical technique for real-time binding analysis

    • Allows high-throughput screening of multiple antibody variants

    • Enables comparative ranking of affinity across a panel of candidates

  • Cell-based binding assays:

    • Measure antibody binding to BR3-expressing cells by flow cytometry

    • Generate saturation binding curves using increasing antibody concentrations

    • Calculate apparent KD values in cellular context

    • Compare binding to cells expressing different levels of BR3

  • Isothermal titration calorimetry (ITC):

    • Measure thermodynamic parameters of binding (ΔH, ΔS, ΔG)

    • Determine stoichiometry of interaction

    • Complement kinetic measurements with equilibrium thermodynamics

How should researchers design experiments to evaluate BR3 antibody effects on B-cell function?

Assessing functional effects of BR3 antibodies requires carefully designed experimental approaches:

  • In vitro B-cell proliferation assay:

    • Isolate human B cells from peripheral blood or tonsil tissue

    • Culture with BAFF to stimulate proliferation

    • Add BR3 antibodies at various concentrations

    • Measure proliferation using methods like 3H-thymidine incorporation or CFSE dilution

    • Compare antagonistic effects of different antibodies (e.g., CB1 variants)

  • B-cell survival assessment:

    • Culture purified B cells in serum-free or low-serum conditions

    • Add BAFF with or without BR3 antibodies

    • Measure viability using Annexin V/PI staining

    • Quantify dose-dependent effects on BAFF-mediated survival

  • Signaling pathway analysis:

    • Stimulate B cells with BAFF in presence/absence of BR3 antibodies

    • Lyse cells and perform Western blotting for phosphorylated signaling proteins

    • Analyze activation of NF-κB, MAP kinase, and other relevant pathways

    • Compare signaling modulation across different antibody variants

  • In vivo B-cell depletion study:

    • Administer BR3 antibodies to mice at varying doses

    • Collect peripheral blood, spleen, and lymph nodes at multiple timepoints

    • Analyze B-cell populations by flow cytometry

    • Assess dose-response and time-course of B-cell reduction

What approach should researchers take when comparing different BR3 antibody clones?

Systematic comparison of BR3 antibody clones requires a multi-parameter assessment:

  • Binding profile analysis:

    • Measure affinity for human and murine BR3 using SPR

    • Compare epitopes using competition assays or epitope mapping

    • Evaluate binding to different B-cell subpopulations by flow cytometry

    • BR3 and BR31 demonstrate distinct staining patterns, with BR31 recognizing only a subpopulation (about 50%) of B lymphocytes

  • Functional comparison methodology:

    • Test each clone in parallel using standardized functional assays

    • Compare dose-response curves for BAFF-blocking activity

    • Assess B-cell depletion potency in vivo

    • Evaluate potential agonistic versus antagonistic effects

  • Stability and manufacturability assessment:

    • Analyze thermal stability using differential scanning calorimetry

    • Evaluate aggregation propensity by size-exclusion chromatography

    • Compare expression yields in production systems

    • Assess long-term stability under various storage conditions

  • Cross-reactivity profiling:

    • Test binding to related receptors (TACI, BCMA)

    • Evaluate reactivity with BR3 from different species

    • Screen against tissue panels to identify potential off-target binding

What methodological considerations are important when developing BR3 antibodies for therapeutic applications?

Translating research-grade BR3 antibodies to therapeutic candidates involves additional methodological considerations:

  • Humanization or human antibody selection strategy:

    • For mouse-derived antibodies like BR3, BR16, and BR31 , humanization is required

    • Alternatively, select fully human antibodies from phage display libraries

    • Confirm that humanization or selection doesn't compromise binding or function

  • Fc engineering approach:

    • Modify Fc region to enhance or reduce effector functions based on therapeutic goal

    • For B-cell depletion, enhance ADCC/CDC through afucosylation or point mutations

    • For blocking applications without cell depletion, consider effector-silent mutations

    • Optimize half-life through FcRn-binding modifications

  • Developability assessment:

    • Evaluate sequence for potential immunogenic T-cell epitopes

    • Screen for chemical stability issues (deamidation, oxidation sites)

    • Assess glycosylation profile and impact on function

    • Test behavior in formulation buffers relevant for therapeutic development

  • Translational model selection:

    • Choose appropriate animal models accounting for BR3 sequence differences

    • Consider using transgenic models expressing human BR3

    • Design studies to assess both efficacy and potential toxicity

    • For bispecific approaches, ensure both targets are properly represented in models

How should researchers analyze and interpret BR3 antibody binding heterogeneity across B-cell populations?

BR3 antibody binding can vary across B-cell subsets, requiring sophisticated analysis approaches:

  • Multi-parameter flow cytometry analysis:

    • Combine BR3 antibody with markers for B-cell subpopulations

    • Design panels to identify naïve, memory, transitional, and plasmablast populations

    • Quantify BR3 expression levels (MFI) across subsets

    • Compare binding patterns of different BR3 antibody clones (e.g., BR3 vs. BR31)

  • Correlation analysis methodology:

    • Calculate correlation coefficients between BR3 staining and other B-cell markers

    • Compare patterns in peripheral blood versus lymphoid tissues

    • Analyze how correlation patterns differ between antibody clones

    • BR3 staining pattern closely correlates with anti-human Ig in both peripheral blood and lymphoid tissues

  • Heterogeneity quantification:

    • Apply clustering algorithms to identify distinct B-cell populations

    • Calculate coefficient of variation in BR3 staining within each population

    • Compare heterogeneity patterns between healthy and disease samples

    • Interpret subset-specific binding in context of B-cell development

  • Visualization approach:

    • Create biaxial plots showing BR3 staining versus other markers

    • Generate heat maps showing relative BR3 expression across all identified subsets

    • Use dimensionality reduction techniques (tSNE, UMAP) for complex datasets

    • Present data showing BR31 recognizes approximately 50% of B lymphocytes

What statistical methods are appropriate for analyzing BR3 antibody affinity and potency data?

Rigorous statistical analysis is essential for meaningful interpretation of BR3 antibody characteristics:

How can researchers leverage antibody sequence databases to guide BR3 antibody optimization?

The wealth of antibody sequence data provides valuable guidance for BR3 antibody engineering:

  • Public CDR-H3 analysis approach:

    • Compare BR3 antibody sequences to public CDR-H3 databases

    • Identify sequence features associated with highly public antibodies (shorter length, lower diversity)

    • Use these insights to guide optimization of therapeutic candidates

    • Consider that only 0.07% of unique CDR-H3s are highly public (occurring in ≥5 bioprojects)

  • Sequence-structure relationship mining:

    • Compare BR3 antibody sequences with structurally characterized antibodies

    • Identify canonical structures for CDRs that may enhance stability or function

    • Apply machine learning methods to predict performance based on sequence features

    • Leverage the AbNGS database containing 4 billion productive human heavy variable region sequences

  • Germline divergence analysis:

    • Calculate amino acid replacement:silent mutation ratios in CDRs vs. frameworks

    • Assess degree of somatic hypermutation relative to germline sequences

    • Consider reverting non-essential mutations to germline to reduce immunogenicity

    • Compare natural vs. synthetic antibody approaches for BR3 targeting

  • Bioinformatic optimization methodology:

    • Use frequency-based approaches to identify favorable residues at each position

    • Apply computational design algorithms to suggest stability-enhancing mutations

    • Predict potential post-translational modification sites that may affect function

    • Design focused libraries targeting key positions for experimental validation

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.