The BCL10 antibody is a specific reagent designed to detect the B-cell lymphoma/leukemia 10 (BCL10) protein, a critical adaptor molecule in immune signaling pathways. BCL10 contains a caspase recruitment domain (CARD) and mediates interactions between upstream signaling molecules (e.g., CARD9, CARD11) and downstream effectors like MALT1, facilitating NF-κB activation and apoptosis . Monoclonal (e.g., clones BSB-22, 4F8) and polyclonal (e.g., Proteintech 17732-1-AP) variants are available, optimized for techniques such as immunohistochemistry (IHC), western blotting (WB), and flow cytometry (FACS) .
Target: BCL10 protein (26 kDa, 233 amino acids).
Applications: IHC, WB, FACS, ELISA, and immunofluorescence (IF) .
BCL10 is encoded by the BCL10 gene on chromosome 1 and consists of an N-terminal CARD domain and a C-terminal Ser/Thr-rich region . It forms filaments that amplify signaling through the CBM (CARMA/CARD-BCL10-MALT1) complex, regulating adaptive and innate immunity .
NF-κB Activation: BCL10 bridges CARD-containing scaffold proteins (e.g., CARD11) to MALT1 protease, enabling NF-κB and JNK signaling .
Apoptosis and Survival: Promotes pro-caspase-9 maturation and apoptosis via interactions with TNFR1-TRADD-RIP complexes .
Immune Regulation: Required for T-cell activation, cytokine production (e.g., IL-2), and development of regulatory T cells .
The antibody is used in both diagnostic and research settings to study BCL10 expression and its role in immune disorders.
BCL10 overexpression is associated with mucosa-associated lymphoid tissue (MALT) lymphomas, where it forms nuclear aggregates due to chromosomal translocations (e.g., t(1;14)) . The antibody aids in:
Lymphoma Classification: Distinguishes MALT lymphomas (55% positivity) from other B-cell malignancies .
Therapeutic Monitoring: Assesses BCL10 degradation in activated T cells, linked to NF-κB signaling defects .
Recent studies highlight BCL10’s dynamic regulation:
Post-Translational Modifications: Phosphorylation, ubiquitination, and MALT1-mediated cleavage modulate its stability and signaling .
Degradation: T-cell activation induces rapid BCL10 proteolysis, impairing NF-κB activation .
Therapeutic Potential: Targeting BCL10 or its interactors (e.g., MALT1) may treat autoimmune diseases or lymphomas .
BCL10 Function: Relevant Research Findings
Applications : Immuno-histochemistry
Sample type: HeLa cells
Sample dilution: 1:1000
Review: For colon cancer immuno-histochemistry, tumour biopsies were formalin fixed and paraffin embedded. Tissue sections were treated with EnVision FLEX. Target retrieval solution low pH in order to unmask the antigens. The immunolabelling was performed in the same way as that for the cultured cells.
What is BCL10 and why is it important in immunological research?
BCL10 is a 26.3 kDa protein (233 amino acids in humans) that functions as an immune signaling adaptor. It plays a crucial role in both adaptive and innate immune signaling by bridging CARD domain-containing proteins to immune activation. BCL10 is essential for proper immune function as it channels signals downstream of CARD domain-containing proteins CARD9, CARD11, and CARD14 to activate NF-κB and MAP kinase p38 pathways, which ultimately stimulate the expression of genes encoding pro-inflammatory cytokines and chemokines .
BCL10 is ubiquitously expressed across many tissue types and has been implicated in various immunological processes. Research on BCL10 is particularly important because:
It is critical for T-cell and B-cell receptor signaling
Human BCL10 deficiency leads to severe immunological disorders
It undergoes key post-translational modifications including ubiquitination, phosphorylation, and protein cleavage
It forms part of the CBM (CARD9/11/14-BCL10-MALT1) signaling complex critical for immune activation
What applications are BCL10 antibodies commonly used for?
BCL10 antibodies are utilized across multiple research applications, with varying protocols and considerations for each:
| Application | Common Usage | Typical Dilutions | Notes |
|---|---|---|---|
| Western Blot (WB) | Most widely used application | 0.04-0.4 μg/mL to 1/500-1/1000 | Detects band at ~26-28 kDa |
| Immunohistochemistry (IHC-P) | Detection in tissue samples | 1:20-1:50 to 1 μg/ml | Works on formalin-fixed paraffin-embedded tissues |
| Immunofluorescence (IF/ICC) | Subcellular localization | 10 μg/mL | Shows primarily cytoplasmic staining |
| Flow Cytometry | Cell population analysis | Varies by antibody | Requires cell permeabilization for intracellular staining |
| Immunoprecipitation (IP) | Protein interaction studies | 2-4 μg per sample | Effective for pull-down of BCL10 complexes |
| ELISA | Quantitative detection | Varies by kit | Available in kit format for human samples |
When selecting BCL10 antibodies for specific applications, researchers should review validation data for the specific application and species of interest .
How should samples be prepared for optimal BCL10 detection by Western blot?
For optimal detection of BCL10 by Western blot, follow these methodological guidelines:
Cell/Tissue Lysis: Use an IP lysis buffer containing protease inhibitors to prevent degradation. For typical experiments, a buffer containing 1% Triton X-100, 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, and a protease inhibitor cocktail is effective .
Sample Processing:
For adherent cells: Wash with PBS, add lysis buffer directly to the plate, scrape, and collect
For suspension cells: Pellet cells, resuspend in lysis buffer
For tissues: Homogenize in lysis buffer using a mechanical homogenizer
Protein Denaturation: Mix with Laemmli buffer (containing SDS and β-mercaptoethanol) and heat at 95°C for 5 minutes.
Gel Selection: Use 10-12% SDS-PAGE gels for optimal resolution of BCL10 (26.3 kDa).
Transfer Conditions: Transfer to PVDF or nitrocellulose membranes at 100V for 1 hour or 30V overnight.
Blocking: Block membranes with 5% non-fat milk or BSA in TBST for 1 hour at room temperature.
Antibody Incubation: Dilute primary antibody according to manufacturer's recommendations (typically 0.04-0.4 μg/mL or 1:500-1:1000) and incubate overnight at 4°C .
Note that BCL10 may undergo degradation during T-cell activation, so sample timing is critical when working with activated immune cells .
How does T-cell activation affect BCL10 protein levels, and how should this be considered in experimental design?
T-cell activation significantly impacts BCL10 protein levels through regulated degradation, which serves as a negative feedback mechanism for NF-κB signaling. This phenomenon must be carefully accounted for in experimental designs:
Mechanism of BCL10 degradation during T-cell activation:
TCR/CD28 costimulation or PMA treatment induces rapid degradation of BCL10 protein
CD3 ligation alone has minimal effect, while CD3/CD28 combined stimulation causes significant BCL10 downregulation within 3 hours
BCL10 degradation occurs via the lysosomal pathway rather than the proteasome pathway
The HECT domain ubiquitin ligases NEDD4 and Itch promote ubiquitination and degradation of BCL10
This degradation serves to attenuate NF-κB signaling as a negative feedback mechanism
Experimental design considerations:
Time-course studies: When analyzing BCL10-dependent signaling, include early time points (before significant degradation) and later time points to capture the degradation phase.
Stimulation conditions: Compare CD3 alone versus CD3/CD28 costimulation to differentiate between partial and complete activation signals.
Inhibitor experiments: Include lysosomal inhibitors (e.g., Bafilomycin) to block degradation if sustained BCL10 levels are needed.
Controls: Include TNF-α stimulated samples as controls, as this stimulus activates NF-κB without triggering BCL10 degradation.
Imaging studies: For microscopy, be aware that BCL10 transiently localizes to lysosomal vesicles during degradation, which may affect interpretation of localization studies .
These considerations are critical for accurately interpreting experiments involving T-cell activation and BCL10-dependent signaling pathways.
What are the key methodological approaches for studying BCL10 protein-protein interactions?
Studying BCL10 protein-protein interactions requires multiple complementary approaches to comprehensively understand its binding partners and complex formation:
Co-immunoprecipitation (Co-IP):
Lyse cells in a buffer containing 1% Triton X-100, 50 mM Tris-HCl (pH 7.4), 150 mM NaCl with protease inhibitors
Immunoprecipitate with 2-4 μg of anti-BCL10 antibody or tag-specific antibody for tagged constructs
Capture with Protein G-Sepharose (1 hour, 4°C)
Wash extensively (3× with IP lysis buffer)
Analyze by SDS-PAGE and Western blot for interacting partners (e.g., MALT1, CARMA1/3, CARD9)
Förster Resonance Energy Transfer (FRET) analysis:
Generate fluorescent protein fusions (CFP/YFP or similar pairs) of BCL10 and potential interaction partners
Express in appropriate cell models (primary cells or cell lines)
Analyze by confocal microscopy or flow cytometry-based FRET
For flow cytometry FRET, use appropriate compensation controls (1.0-1.2% for FRET-YFP and 80-90% for FRET-CFP)
For microscopy-based FRET, fluorescence lifetime imaging microscopy (FLIM) provides quantitative measurement of protein interactions
Mutagenesis approaches:
Generate deletion mutants to map interaction domains
For BCL10-MALT1 interactions, focus on residues 107-119 of BCL10, which are critical for binding
Create point mutations in conserved, solvent-exposed residues identified through structural modeling
Test interactions using reporter assays (e.g., NF-κB luciferase assay) and co-IP experiments
Validate in cellular contexts using reconstitution of BCL10-deficient cells
These approaches can reveal critical insights into how BCL10 forms complexes with MALT1 and CARD domain-containing proteins to regulate immune signaling.
How can researchers distinguish between different BCL10 post-translational modifications?
BCL10 undergoes multiple post-translational modifications (PTMs) including phosphorylation, ubiquitination, and proteolytic cleavage. Distinguishing between these modifications requires specific methodological approaches:
Phosphorylation analysis:
Phospho-specific antibodies: While limited commercial phospho-specific BCL10 antibodies exist, researchers can generate custom antibodies against known phosphorylation sites.
Phosphatase treatment: Treat half the sample with lambda phosphatase before Western blotting; shifts in migration patterns indicate phosphorylation.
Phos-tag™ SDS-PAGE: This technique specifically retards the migration of phosphorylated proteins, allowing separation of phosphorylated from non-phosphorylated forms.
Mass spectrometry: For detailed phosphorylation site mapping, immunoprecipitate BCL10 and analyze by LC-MS/MS.
Kinase inhibitors: Use protein kinase C inhibitors (e.g., Bisindolylmaleimide) to block phosphorylation events triggered during T-cell activation .
Ubiquitination analysis:
His-tagged ubiquitin pulldown: Express His-tagged ubiquitin in cells, perform nickel affinity purification under denaturing conditions, and detect BCL10 by Western blot.
Immunoprecipitation with ubiquitin antibodies: IP with anti-ubiquitin antibodies and probe for BCL10.
Proteasome inhibitors: Compare samples treated with MG132 (proteasomal inhibitor) versus lysosomal inhibitors (E64d, Bafilomycin) to distinguish between degradation pathways .
Proteolytic cleavage:
Size analysis: Detect cleavage products using Western blot with antibodies targeting different epitopes of BCL10.
Protease inhibitors: Use specific protease inhibitors to block cleavage and identify responsible proteases.
N-terminal antibodies versus C-terminal antibodies: Compare staining patterns to identify retained portions .
When designing experiments, consider that these modifications often occur in response to specific stimuli and may be transient.
What are the implications of BCL10 deficiency for immune function and how can BCL10 antibodies help study this condition?
BCL10 deficiency represents a primary immunodeficiency with severe clinical manifestations. BCL10 antibodies serve as critical tools for diagnosing and studying this condition:
Clinical manifestations of BCL10 deficiency:
Combined immunodeficiency affecting both T and B cell functions
Recurrent bacterial and viral infections
Impaired memory B cell differentiation
Reduced central memory, effector memory, and TEMRA CD4+ T cell compartments
Hematopoietic stem cell transplantation (HSCT) is the recommended treatment
Methodological approaches using BCL10 antibodies:
Diagnostic immunoblotting: Western blot analysis of PBMCs using anti-BCL10 antibodies to confirm absence of BCL10 protein. This requires sensitive antibodies that can detect very low protein levels .
Flow cytometry with mass cytometry (CyTOF):
Genetic confirmation:
Functional assays:
This combined approach allows for comprehensive analysis of immune defects resulting from BCL10 deficiency and can guide treatment decisions.
How should researchers design validation experiments for new BCL10 antibodies?
Thorough validation of BCL10 antibodies is essential to ensure experimental reliability. A comprehensive validation approach should include:
1. Specificity Validation:
Western blot with positive and negative controls:
Peptide competition assays:
Pre-incubate antibody with immunizing peptide
Compare detection with and without peptide blocking
Signal should be eliminated or significantly reduced with blocking peptide
Cross-species reactivity:
2. Application-specific Validation:
For immunohistochemistry:
For flow cytometry:
For immunoprecipitation:
3. Technical Validation:
Reproducibility testing:
Test antibody performance across different lots
Evaluate consistency across different sample types
Determine stability under various storage conditions
Epitope mapping:
Proper validation ensures reliable results and aids in the interpretation of experimental outcomes when working with BCL10.
What controls should be included when using BCL10 antibodies in signal transduction studies?
When studying BCL10's role in signal transduction pathways, incorporating appropriate controls is essential for reliable data interpretation:
Positive controls:
Stimulation controls: Include samples treated with known activators of BCL10-dependent pathways:
Expression controls: Include cells transfected with BCL10 expression vectors to serve as positive controls for antibody reactivity
Negative controls:
Pathway-specific controls: Include TNF-α stimulated samples which activate NF-κB independent of BCL10
Cell-type controls: Include cell lines lacking BCL10 expression or with BCL10 knocked down via RNAi
Domain-specific controls: Use BCL10 mutants with specific domain deletions:
Time-course controls:
Include multiple time points after stimulation (e.g., 0, 15, 30, 60, 120, 180 minutes) to capture the dynamic regulation of BCL10, including its degradation following T-cell activation
Inhibitor controls:
Pathway inhibitors:
Loading controls: Always include appropriate loading controls:
Including these controls allows for proper interpretation of BCL10's role in signaling cascades and helps distinguish between specific effects and experimental artifacts.
How can researchers optimize immunohistochemistry protocols for BCL10 detection in different tissue types?
Optimizing immunohistochemistry (IHC) protocols for BCL10 detection requires tissue-specific considerations and careful method development:
General Protocol Optimization:
Antigen Retrieval Methods:
Antibody Selection and Dilution:
Signal Detection Systems:
Compare polymer-based detection vs. avidin-biotin complex (ABC) methods
For low-expression tissues, consider signal amplification systems
Optimize DAB development time for specific antibodies and tissues
Tissue-Specific Considerations:
| Tissue Type | Special Considerations | Recommended Protocol Modifications |
|---|---|---|
| Lymphoid Tissues | High endogenous BCL10 expression | Lower antibody concentration (1:50-1:100), shorter incubation times |
| Tonsil | Excellent positive control tissue | Standard protocol serves as reference point |
| Gastrointestinal | Potential cross-reactivity with gut flora | Increase washing steps, consider lower antibody concentration |
| Brain | Low expression, high background | Extend blocking time, use specialized blocking reagents |
| FFPE vs. Frozen | Different fixation affects epitope accessibility | FFPE typically requires stronger antigen retrieval |
Validation Approaches:
Multiplex staining with lineage markers:
Co-stain with T-cell (CD3) or B-cell (CD20) markers to confirm cell-type specific expression
Use serial sections for comparison if multiplex is not available
RNA-protein correlation:
Compare IHC results with in situ hybridization or RNA-seq data from the same tissue types
This verifies that protein expression patterns match transcript expression
Quantification methods:
These optimization strategies ensure reliable and reproducible BCL10 detection across different tissue types and experimental conditions.
What are common issues with BCL10 antibodies in Western blot applications and how can they be resolved?
Western blotting with BCL10 antibodies can present several technical challenges. Here's a systematic approach to troubleshooting common issues:
| Possible Causes | Solutions |
|---|---|
| Low BCL10 expression | Use positive control lysates (Jurkat, Raji cells) |
| Insufficient antibody concentration | Increase antibody concentration or incubation time |
| Protein degradation | Add protease inhibitors to lysis buffer; maintain samples at 4°C |
| Inefficient transfer | Optimize transfer conditions; verify with Ponceau S staining |
| BCL10 degradation after stimulation | Check time points carefully; BCL10 levels decrease after T-cell activation |
| Possible Causes | Solutions |
|---|---|
| Post-translational modifications | Compare with and without phosphatase treatment |
| Proteolytic fragments | Add additional protease inhibitors; reduce sample processing time |
| Non-specific binding | Increase blocking time/concentration; try different blocking agents |
| Alternative splice variants | Verify with different antibodies targeting different epitopes |
| Cross-reactivity | Test antibody on BCL10-knockout samples as negative control |
| Possible Causes | Solutions |
|---|---|
| Insufficient blocking | Increase blocking time or concentration (5% milk/BSA for 1-2 hours) |
| Excessive antibody concentration | Perform antibody titration to determine optimal concentration |
| Inadequate washing | Increase number and duration of wash steps (5×5 minutes) |
| Membrane issues | Try different membrane types (PVDF vs. nitrocellulose) |
| Secondary antibody problems | Test secondary antibody alone; consider using different type |
Technical Optimization Tips:
Sample preparation optimization:
Gel percentage optimization:
Special considerations for stimulated samples:
Antibody selection strategies:
These approaches should resolve most common issues encountered when using BCL10 antibodies for Western blotting applications.
How can researchers use BCL10 antibodies to investigate the formation and regulation of the CBM signalosome?
The CBM (CARD9/11/14-BCL10-MALT1) signalosome is a critical complex in immune signaling. BCL10 antibodies are invaluable tools for investigating its formation, regulation, and function:
Experimental approaches for studying CBM complex formation:
Sequential co-immunoprecipitation (Co-IP):
Size-exclusion chromatography with antibody detection:
Separate cell lysates by size using gel filtration
Analyze fractions by Western blot using anti-BCL10 antibodies
Compare fractionation profiles before and after cell stimulation
BCL10 shifts to higher molecular weight fractions upon CBM complex formation
FRET-based approaches:
Visualizing CBM signalosome dynamics:
Immunofluorescence microscopy with BCL10 antibodies:
Proximity ligation assay (PLA):
Use antibody pairs targeting different CBM components
PLA signal indicates close proximity (<40 nm)
Quantify PLA signals before and after cell stimulation
This approach provides spatial information about complex formation in situ
Investigating regulatory mechanisms:
Post-translational modification analysis:
Structural requirements for CBM formation:
Use BCL10 antibodies to validate expression of mutant constructs
Key regions to examine include:
BCL10 CARD domain (aa 13-119)
MALT1 binding region (aa 107-119)
Helices 5-6 face containing conserved residues (D80, E84, K90, D101)
Confirm proper folding of mutant constructs using conformation-specific antibodies
Manipulation of CBM complex with inhibitors:
These approaches enable comprehensive investigation of the CBM signalosome's role in immune signaling and potential targeting for therapeutic purposes.
How are BCL10 antibodies being used in studies of BCL10 deficiency and immunodeficiency disorders?
BCL10 deficiency represents a rare but severe primary immunodeficiency. BCL10 antibodies are playing crucial roles in advancing our understanding of this condition:
Diagnostic applications:
Confirmation of BCL10 deficiency:
Carrier detection:
Immunological profiling:
Mass cytometry (CyTOF) with BCL10 antibodies:
Functional assessments:
Emerging research applications:
Gene therapy approaches:
BCL10 antibodies for validating gene correction strategies
Monitoring BCL10 expression after genetic interventions
Correlating restored expression with functional recovery
Partial deficiency studies:
Tissue-specific consequences:
Immunohistochemistry to examine BCL10 expression in tissues
Studying non-immune manifestations of BCL10 deficiency
Correlating tissue-specific expression with clinical phenotypes
This research is providing critical insights into human BCL10 biology and guiding the development of therapeutic strategies for affected patients, particularly the use of hematopoietic stem cell transplantation (HSCT) as a definitive treatment .
What are the latest methodological advances in studying BCL10 interactions with MALT1 and CARD proteins?
Recent technological advances have enabled more sophisticated analysis of BCL10's interactions with its binding partners. These cutting-edge approaches include:
Structural biology approaches:
Cryo-electron microscopy (Cryo-EM):
Visualizes filamentous assemblies formed by BCL10 and its partners
Requires specialized sample preparation and antibody-based validation
Reveals molecular architecture of CBM complexes
BCL10 antibodies can be used to validate protein identity in reconstructed structures
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Maps interaction interfaces between BCL10 and its binding partners
Requires careful validation with mutational studies
BCL10 antibodies confirm protein expression for mutational validation
Computational modeling with experimental validation:
Advanced protein-protein interaction methods:
Fluorescence lifetime imaging microscopy (FLIM-FRET):
Flow cytometry-based FRET:
Bioluminescence resonance energy transfer (BRET):
Alternative to FRET using luciferase-tagged BCL10
Reduces background and photodamage
Antibodies validate expression levels of fusion proteins
Live-cell imaging techniques:
Lattice light-sheet microscopy:
Allows 3D visualization of BCL10-containing complexes in living cells
Reduced phototoxicity enables long-term imaging
Can be validated with fixed-cell antibody staining
Super-resolution microscopy (STORM/PALM):
Nanoscale visualization of BCL10-containing complexes
Antibody-based detection with specialized fluorophores
Reveals spatial organization beyond diffraction limit
Proximity-based protein interaction analysis:
BioID and TurboID:
Fusion of biotin ligase to BCL10 biotinylates proximal proteins
Mass spectrometry identifies interaction partners
Antibodies validate expression of fusion proteins and detect specific interactions
APEX2 proximity labeling:
Electron microscopy-compatible approach for mapping interaction networks
Spatial resolution of BCL10 interaction partners
Antibodies confirm expression of fusion proteins
These advanced methodologies provide unprecedented insights into the molecular mechanisms governing BCL10's interactions with MALT1 and CARD proteins in immune signaling pathways.
How do different BCL10 antibody epitopes affect detection of protein complexes and modified forms?
The epitope specificity of BCL10 antibodies significantly impacts their utility for detecting various protein states and complexes. Understanding these differences is crucial for experimental design and data interpretation:
Epitope mapping considerations:
| Antibody Target Region | Advantages | Limitations | Best Applications |
|---|---|---|---|
| N-terminal CARD domain (aa 1-106) | Detects full-length protein | May miss C-terminal cleavage products | Structural studies of CARD-CARD interactions |
| C-terminal region (aa 107-233) | Detects most cleavage products | May miss N-terminal fragments | Protein stability and degradation studies |
| MALT1 binding region (aa 107-119) | Critical for functional studies | May be blocked in protein complexes | Unbound BCL10 detection |
| Central region (aa 100-200) | Less affected by terminal modifications | May miss some PTM effects | General BCL10 detection |
Impact on detecting protein complexes:
CBM complex detection challenges:
Antibodies targeting the CARD domain may show reduced binding when BCL10 is engaged in CARD-CARD interactions
Epitopes in the MALT1-binding region (aa 107-119) may be masked when BCL10 is bound to MALT1
Solution: Use antibodies targeting multiple epitopes or regions unlikely to be involved in protein interactions
Optimization strategies for complex detection:
Detection of modified BCL10 forms:
Phosphorylated BCL10:
Epitopes containing phosphorylation sites may show reduced antibody binding
Phosphorylation-induced conformational changes can affect epitope accessibility
Western blot may show mobility shifts that affect apparent molecular weight
Solution: Use antibodies targeting regions unlikely to be phosphorylated or phospho-specific antibodies
Ubiquitinated BCL10:
Cleaved BCL10:
Application-specific recommendations:
For studying BCL10 degradation: Use antibodies targeting stable epitopes less affected by post-translational modifications
For CARD-domain interactions: Use antibodies targeting the C-terminal region to avoid interference with CARD-CARD binding
For MALT1 binding studies: Use antibodies targeting the N-terminal CARD domain to avoid the MALT1 binding region
For general BCL10 detection: Select antibodies with epitopes in stable, central regions of the protein
Understanding these epitope considerations enables researchers to select appropriate antibodies for specific experimental questions and correctly interpret their results.