BPC2 Antibody

Shipped with Ice Packs
In Stock

Description

Target Biology: BDCA2 Receptor

BDCA2 is a type II C-lectin receptor exclusively expressed on human pDCs . Key features:

  • Structure: Single extracellular carbohydrate recognition domain, transmembrane region, and short cytoplasmic tail requiring FcεRIγ adaptor for signaling

  • Function: Ligand binding inhibits TLR7/9-mediated interferon-alpha (IFN-α) production via SYK-BTK-PLCγ2 signaling cascade

  • Therapeutic rationale: Overactive pDCs drive IFN-I pathology in autoimmune diseases like systemic lupus erythematosus (SLE)

Anti-BDCA2 Monoclonal Antibody Engineering

Two clinically studied anti-BDCA2 mAbs demonstrate distinct mechanisms:

Property24F4A BIIB059
Ig SubclassHumanized IgG1Humanized IgG1
EpitopeNon-linear conformational epitopeUndisclosed (non-competing with 2D6)
Internalization EC500.017 μg/mL1 μg/mL threshold for recovery
Key MechanismDual CD32a/BDCA2 internalization Sustained BDCA2 downmodulation
PD EffectInhibits 90% IFNα in vitro 70% skin MxA reduction in SLE

Phase I Data for BIIB059 (NCT02525016)

  • BDCA2 Internalization: >90% receptor occupancy within 1 hr post-dose persisting ≥112 days at 20 mg/kg

  • pDC Counts: Transient 50% reduction with full recovery despite continued receptor modulation

  • IFN Pathway Suppression:

    • 70% decrease in cutaneous MxA expression (SLE patients, Week 4)

    • Dose-dependent inhibition of IRG scores in blood (max 40% reduction)

Preclinical Efficacy of 24F4A

  • IFNα Inhibition: IC50 = 0.02 μg/mL in human PBMCs (correlates with EC50 for BDCA2 internalization, R²=0.68)

  • Synergistic Effects: 85% greater suppression of SLE immune complex-induced IFN vs anti-CD32a alone

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
BPC2 antibody; BBR antibody; At1g14685 antibody; F10B6.5 antibody; T5E21.17Protein BASIC PENTACYSTEINE2 antibody; AtBPC2 antibody; GAGA-binding transcriptional activator BBR/BPC2 antibody
Target Names
BPC2
Uniprot No.

Target Background

Function
This antibody targets a transcriptional regulator that specifically binds to GA-rich elements (GAGA-repeats) present in regulatory sequences of genes involved in developmental processes.
Database Links

KEGG: ath:AT1G14685

STRING: 3702.AT1G14685.1

UniGene: At.15024

Protein Families
BBR/BPC family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in seedlings, leaves and pistils. Detected in the base of flowers and tips of carpels, in sepal and petal vasculature, in pollen grains, in young rosette, in the lateral and tip of primary roots, and in ovule at the exception of the outer integu

Q&A

What is BDCA2 and why are antibodies against it significant in research?

BDCA2 (Blood Dendritic Cell Antigen 2) is a plasmacytoid dendritic cell (pDC)-specific receptor that, when engaged, inhibits the production of type I interferons (IFN-I) in human pDCs . This makes it an attractive therapeutic target for inhibiting pDC-derived IFN-I, which is implicated in the pathogenesis of autoimmune disorders such as systemic lupus erythematosus (SLE) . Antibodies against BDCA2 are significant because they can engage this receptor and lead to its internalization, consequently inhibiting TLR-induced IFN-I production by pDCs . This inhibitory mechanism makes anti-BDCA2 antibodies valuable tools for both research into pDC biology and potential therapeutic development.

How can researchers verify the specificity of anti-BDCA2 antibodies?

Verifying antibody specificity is crucial for reliable research outcomes. Researchers should implement a multi-step verification approach:

  • Test the antibody on BDCA2-positive cells (pDCs) and BDCA2-negative cells to confirm binding specificity

  • Use flow cytometry to analyze cell surface BDCA2 levels on pDCs (CD14-CD20-HLA-DR+CD123+ cells) before and after antibody treatment

  • Compare results with non-cross-blocking anti-BDCA2 antibodies like clone 2D6 as controls

  • Perform internalization assays in whole blood and isolated pDCs to confirm functional activity

  • Include isotype controls to rule out non-specific binding

Remember that approximately 50% of commercial antibodies fail to meet basic standards for characterization, resulting in significant financial losses and research inconsistencies . Therefore, thorough validation is essential before proceeding with extensive experimentation.

What controls should be included in experiments using anti-BDCA2 antibodies?

Proper controls are essential when working with anti-BDCA2 antibodies:

Control TypePurposeExample
Isotype ControlControls for non-specific bindingHuman IgG1
Unrelated Target ControlControls for general antibody effectsAnti-CD40 antibody
Concentration SeriesEstablishes dose-response10, 1, 0.1, 0.001, 0.0001 μg/ml
Cell Type ControlConfirms target specificityTest on non-pDC populations
Blocking ControlConfirms mechanismAnti-CD32 (AT10)

Additionally, when using anti-BDCA2 antibodies to inhibit IFN-I production, researchers should include stimulation controls (such as CpG-A or R848) to verify the inhibitory effect . These controls are critical because the issues with antibody quality are often compounded by insufficient training in antibody use and inadequate control experiments .

How do effector-competent versus effectorless anti-BDCA2 antibodies differ in their mechanisms and efficacy?

Anti-BDCA2 antibodies can be designed with different Fc regions that significantly impact their mechanisms of action and therapeutic potential:

Effector-competent anti-BDCA2 antibodies (like 24F4A with human wild-type IgG1) utilize a dual mechanism of action . First, they engage BDCA2 directly, leading to receptor internalization and inhibition of TLR-induced IFN-I production. Second, their Fc region plays a critical role in potent inhibition of immune complex (IC)-induced IFN-I production through the internalization of CD32a (FcγRIIa) . This dual mechanism potentially enhances clinical efficacy in conditions like SLE.

In contrast, effectorless forms (like 24F4A-ef, a human IgG4.P/human IgG1 chimeric) engage BDCA2 but lack the potent Fc-mediated effects . This difference is particularly important when targeting IC-mediated activation of pDCs, as seen in autoimmune conditions. In SLE, immune complexes bind to CD32a on pDCs and stimulate IFN-I secretion; effector-competent antibodies can interfere with this pathogenic mechanism more effectively than effectorless variants .

Researchers investigating therapeutic applications should consider these mechanistic differences when selecting antibody formats for their studies.

What is the relationship between BDCA2 antibodies and the treatment of systemic lupus erythematosus (SLE)?

Anti-BDCA2 antibodies represent a promising therapeutic approach for SLE treatment based on their ability to target key pathogenic mechanisms:

In SLE pathogenesis, immune complexes bind to the CD32a receptor on pDCs and stimulate excessive secretion of type I interferons, which are strongly implicated in disease development . Anti-BDCA2 antibodies, particularly humanized monoclonal antibodies like 24F4A, can inhibit this process through two complementary mechanisms:

  • Direct engagement of BDCA2, causing receptor internalization and inhibition of TLR-induced IFN-I production

  • Fc-mediated internalization of CD32a, preventing immune complex binding and subsequent pDC activation

Research has demonstrated that 24F4A inhibits pDC activation by SLE-associated immune complexes and effectively reduces IFN-I production in blood samples from both healthy and SLE donors . This inhibitory effect was confirmed in vivo using a single injection of 24F4A in cynomolgus monkeys .

The therapeutic potential of anti-BDCA2 antibodies is particularly relevant for SLE patients with high interferon signatures, recurring thrombotic events, or pregnancy complications, which are all linked to the overproduction of type I interferons .

How do BDCA2 antibodies interact with immune complexes and modulate CD32a function?

Anti-BDCA2 antibodies demonstrate a sophisticated interaction with immune complexes (IC) and CD32a receptors on plasmacytoid dendritic cells:

In SLE pathology, immune complexes containing auto-antigens like Sm/RNP bind to CD32a (FcγRIIa) on pDCs, triggering IFN-I production . Effector-competent anti-BDCA2 antibodies like 24F4A counter this pathogenic process through a dual mechanism:

  • Direct BDCA2 engagement: Binding to BDCA2 triggers receptor internalization and inhibits TLR-mediated signaling

  • Fc-dependent CD32a modulation: The Fc region of these antibodies is critical for potent inhibition of IC-induced IFN-I production through internalization of CD32a

Experimental evidence demonstrates this mechanism through assays where immune complexes are pre-formed by mixing Sm/RNP antigen with anti-RNP antibodies . When pDCs are treated with these complexes, they produce IFN-I, but this production is inhibited by anti-BDCA2 antibodies. Notably, this inhibition is significantly more potent with effector-competent antibodies compared to effectorless variants, highlighting the importance of the Fc region in therapeutic efficacy .

The internalization of both BDCA2 and CD32a can be quantified through flow cytometry, providing a method to assess the potency of different anti-BDCA2 antibody formulations .

What are the optimal protocols for evaluating anti-BDCA2 antibody efficacy in inhibiting IFN-I production?

Researchers should follow these optimized protocols when evaluating anti-BDCA2 antibody efficacy:

For PBMC-based assays:

  • Isolate PBMCs using Ficoll gradients (overlay whole blood onto Ficoll and centrifuge for 20 min at 600g with no brake)

  • Plate 1 × 10^6 cells/well in complete RPMI media (10% FBS, 1× non-essential amino acids, 1× Pen-Strep, 1 mM sodium pyruvate, 10 mM HEPES, 50 μM 2-mercaptoethanol, 2 mM L-glutamine) in 96-well round-bottom plates

  • Treat with anti-BDCA2 antibodies using a concentration series (10, 3.33, 1.11, 0.37, 0.124, 0.04, 0.014, 0.005, 0.0015, 0.005 μg/ml) or 10 μg/ml of isotype control

  • Stimulate cells with 1 μM CpG-A or 50 μg/ml poly(I:C)

  • Culture for 16 hours at 37°C and 5% CO2

  • Collect supernatants and evaluate IFNα levels using a validated ELISA kit (such as VeriKine™ Human Interferon Alpha ELISA)

For isolated pDC assays:

  • Isolate pDCs (CD14-CD20-HLA-DR+CD123+ cells)

  • Plate at 1 × 10^5 cells/well in complete RPMI media

  • Treat with anti-BDCA2 antibodies at various concentrations (10, 1, 0.1, 0.001, 0.0001 μg/ml)

  • Stimulate with 1 μM CpG-A or 5 μM R848

  • Culture and analyze as above

For immune complex stimulation:

  • Pre-form immune complexes by mixing 1.25 μl Sm/RNP antigen and 2.5 μl anti-RNP antibodies, incubating for 30 minutes at room temperature

  • Dilute the mixture in media and add to cells

  • Treat with anti-BDCA2 antibodies or 10 μg/ml anti-CD32 as a control

  • Follow culture and analysis steps as above

Set up all conditions in duplicate and calculate average IFNα levels for accurate results .

How can researchers accurately measure BDCA2 receptor internalization following antibody binding?

Measuring BDCA2 receptor internalization requires precise techniques:

For whole blood internalization assays:

  • Treat whole blood with anti-BDCA2 antibodies using a concentration series (10 to 0.005 μg/ml) at 37°C for various time points

  • Stain with antibodies against cell surface markers: anti-CD123 (7G3), anti-CD20 (2H7), anti-CD14 (M5E2), anti-HLADR (L243), and a non-cross-blocking anti-BDCA2 clone (2D6)

  • Lyse red blood cells using BD lyse/fix solution according to the manufacturer's protocol

  • Define pDCs as CD14-CD20-HLA-DR+CD123+ cells

  • Use flow cytometry to evaluate remaining surface BDCA2 levels on pDCs

For isolated pDC internalization assays:

  • Treat isolated pDCs with anti-BDCA2 antibodies at various concentrations

  • Include appropriate controls (anti-CD40 antibody or isotype control)

  • After treatment, stain cells with anti-BDCA2 (clone 2D6) and anti-CD32 (AT10)

  • Acquire data on a flow cytometer (LSRII or Fortessa) and analyze using FlowJo software

The decrease in detectable surface BDCA2 directly correlates with receptor internalization efficacy. This can be quantified as percent reduction in mean fluorescence intensity compared to untreated controls. Time-course experiments are particularly valuable for determining the kinetics of receptor internalization.

What factors should researchers consider when selecting anti-BDCA2 antibodies for specific experimental applications?

When selecting anti-BDCA2 antibodies, researchers should carefully evaluate:

  • Antibody format and Fc region: Determine whether your application requires effector-competent (wild-type IgG1) or effectorless (IgG4.P/IgG1 chimeric) antibodies . Effector-competent antibodies provide enhanced inhibition of immune complex-induced responses through CD32a internalization .

  • Characterization status: Verify that the antibody has undergone rigorous characterization. With approximately 50% of commercial antibodies failing to meet basic standards, proper characterization is essential for reliable results .

  • Application-specific validation: Confirm that the antibody has been validated for your specific application (flow cytometry, ELISA, immunoprecipitation, etc.) .

  • Clone selection: Different clones may have different epitope specificities and functional effects. For example, clone 24F4A has been specifically developed and validated for inhibiting pDC activation .

  • Cross-reactivity and species specificity: Confirm that the antibody works in your experimental system. Many anti-BDCA2 antibodies are human-specific and may not cross-react with other species .

  • Controls availability: Ensure you can obtain appropriate control antibodies (isotype controls, non-cross-blocking anti-BDCA2 clones for detection) .

  • Reproducibility data: Review available literature and vendor data on lot-to-lot consistency and reproducibility .

Remember that inadequate antibody characterization costs the research community billions of dollars annually in wasted experiments and irreproducible results . Taking time to properly select and validate antibodies will significantly improve research quality and reliability.

How do beta-2 glycoprotein 1 antibodies differ from BDCA2 antibodies in their clinical applications?

While both are antibodies with potential therapeutic applications, beta-2 glycoprotein 1 antibodies and BDCA2 antibodies target different biological systems and have distinct clinical applications:

Beta-2 glycoprotein 1 antibodies:

  • Are autoantibodies associated with inappropriate blood clotting

  • Target the body's own lipid-proteins found in cell membranes and platelets

  • Are associated with antiphospholipid syndrome (APS), characterized by widespread blood clots, low platelet count, and pregnancy complications

  • Are primarily used diagnostically to help identify APS, unexplained blood clots, and recurrent miscarriages

  • May be detected in three different classes (IgG, IgM, and IgA), with IgG and IgM being most clinically relevant

BDCA2 antibodies:

  • Are therapeutic antibodies targeting a receptor on plasmacytoid dendritic cells

  • Function by inhibiting type I interferon production implicated in autoimmune diseases

  • Have potential applications in treating systemic lupus erythematosus (SLE) and other interferon-driven autoimmune conditions

  • Work through a dual mechanism involving receptor internalization and in some cases, Fc-mediated effects

  • Are still primarily in research and development stages rather than established diagnostic tools

These fundamental differences make these antibodies suitable for distinct clinical scenarios: beta-2 glycoprotein 1 antibodies for diagnosing thrombotic disorders and pregnancy complications, and BDCA2 antibodies for potential treatment of interferon-driven autoimmune diseases.

What research challenges remain in developing anti-BDCA2 antibodies as therapeutic agents?

Despite promising results, several challenges persist in developing anti-BDCA2 antibodies as therapeutic agents:

  • Determining optimal antibody format: Research must clarify whether effector-competent or effectorless antibodies provide the best therapeutic profile in different patient populations . While effector-competent antibodies show enhanced inhibition of immune complex-induced interferon production, the potential for unwanted effector functions must be evaluated .

  • Establishing dosing regimens: Current research has primarily examined single-dose effects in animal models . Determining optimal dosing frequency, concentration, and duration for sustained clinical benefit remains challenging.

  • Predicting long-term effects: The long-term consequences of BDCA2 engagement and pDC modulation require further investigation. Potential compensatory mechanisms or resistance could develop with prolonged treatment.

  • Patient stratification: Identifying which patients would benefit most from anti-BDCA2 therapy is crucial. This likely requires developing biomarkers for interferon signature and pDC activity.

  • Antibody characterization standardization: With the broader issue of inadequate antibody characterization in biomedical research , establishing standardized protocols for characterizing therapeutic anti-BDCA2 antibodies is essential for consistent manufacturing and regulatory approval.

  • Balancing immune suppression: While inhibiting interferon production may benefit autoimmune conditions, potential increased susceptibility to viral infections must be carefully assessed.

  • Combination therapy approaches: Research into combining anti-BDCA2 antibodies with other immunomodulatory agents could enhance efficacy but adds complexity to development and safety assessment.

Addressing these challenges requires coordinated efforts between academic researchers, pharmaceutical companies, regulatory agencies, and clinical investigators to advance these promising therapeutics from bench to bedside.

How should researchers design experiments to study the effects of anti-BDCA2 antibodies on immune complex-mediated pDC activation?

When studying immune complex-mediated pDC activation and its inhibition by anti-BDCA2 antibodies, researchers should implement the following experimental design:

Immune Complex Preparation:

  • Reconstitute anti-RNP antibodies in water

  • Pre-form immune complexes by mixing Sm/RNP antigen (1.25 μl) with anti-RNP antibodies (2.5 μl) and incubating for 30 minutes at room temperature

  • Dilute the mixture in media before adding to cells

Experimental Groups:

  • Positive control: pDCs with immune complexes alone

  • Treatment groups: pDCs with immune complexes plus anti-BDCA2 antibodies at various concentrations

  • Mechanism control: pDCs with immune complexes plus anti-CD32 (AT10) at 10 μg/ml

  • Format comparison: Include both effector-competent (e.g., 24F4A) and effectorless (e.g., 24F4A-ef) anti-BDCA2 antibodies to compare their effects

  • Negative control: pDCs without immune complex stimulation

Readouts:

  • Primary: IFNα production measured by ELISA

  • Secondary:

    • Surface BDCA2 levels by flow cytometry

    • CD32a receptor internalization

    • pDC activation markers

    • Cell viability assessments

Analysis Approaches:

  • Calculate percent inhibition of IFNα production relative to positive control

  • Determine IC50 values for different antibody formats

  • Correlate BDCA2/CD32a internalization with inhibition of IFNα production

  • Compare the potency of effector-competent versus effectorless antibodies in this specific context

This comprehensive experimental design will provide insights into both the efficacy and mechanisms of anti-BDCA2 antibodies in countering immune complex-mediated pDC activation, a key pathogenic process in autoimmune diseases like SLE.

What are the best practices for antibody characterization to ensure reproducible results?

To ensure reproducible results with antibodies, including anti-BDCA2 antibodies, researchers should follow these best practices for characterization:

  • Validate specificity through multiple approaches:

    • Test binding to target-expressing and non-expressing cells

    • Use western blots or immunoprecipitation followed by mass spectrometry

    • Employ genetic approaches (knockout/knockdown) to confirm specificity

    • Compare results across multiple antibody clones targeting different epitopes

  • Determine optimal working conditions:

    • Establish concentration range through dose-response curves (e.g., 10 μg/ml to 0.0001 μg/ml)

    • Test in relevant biological matrices (whole blood, isolated cells, tissues)

    • Optimize incubation time, temperature, and buffer conditions

  • Document comprehensive metadata:

    • Record complete antibody information: clone, lot number, manufacturer

    • Specify exact experimental conditions used for validation

    • Document all positive and negative controls employed

  • Perform functional validation:

    • For anti-BDCA2 antibodies, assess internalization capacity

    • Measure inhibition of type I interferon production in response to TLR stimulation

    • Test efficacy against immune complex-induced activation

  • Assess reproducibility:

    • Test multiple antibody lots

    • Repeat experiments on different days and by different researchers

    • Verify results across different biological samples/donors

  • Share characterization data:

    • Deposit validation data in public repositories

    • Include detailed methods sections in publications

    • Report negative results and limitations

Implementing these practices addresses the "antibody characterization crisis" that costs the research community billions of dollars annually in wasted experiments and irreproducible results . Remember that approximately 50% of commercial antibodies fail to meet even basic characterization standards, making rigorous validation essential before conducting extensive experiments .

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.