Western Blot: Detects RelB at ~70 kDa in lysates from Burkitt’s lymphoma cell lines (Daudi, Raji) .
Immunohistochemistry: Localizes RelB in paraffin-embedded human lymphoma tissues at 8 µg/mL .
Immunofluorescence: Confirms cytoplasmic RelB localization in Raji cells using 25 µg/mL antibody .
NF-κB Pathway Analysis: RelB-p50/p52 heterodimers activate transcription of genes linked to immune regulation and cancer .
Knockdown Validation: siRNA-mediated RelB silencing abolishes antibody binding, confirming specificity .
Quality Control (QC): Used as a control reagent in rapid diagnostic kits to validate antigen integrity .
Gold Nanoparticle Conjugation: Enables lateral flow assay development for antigen detection .
Recombinant antibodies show enhanced sensitivity vs. traditional monoclonals in kinetochore detection .
Engineered light/heavy chains improve signal-to-noise ratios in multiplex assays .
The RELB recombinant monoclonal antibody production is a meticulously controlled process. It begins with in vitro cloning, where the genes encoding both the heavy and light chains of the RELB antibody are inserted into plasmid vectors. These recombinant vectors are then introduced into host cells, providing an environment for the expression of the recombinant antibody within a cell culture. Following expression, the RELB recombinant monoclonal antibody is purified from the supernatant of transfected host cell lines using affinity chromatography. This antibody exhibits high specificity in binding to the human RELB protein. Its versatility allows it to be used in a wide range of applications, including ELISA, immunofluorescence (IF), and flow cytometry (FC).
RELB is a member of the Rel/NF-κB family of transcription factors, which play critical roles in regulating various cellular processes, particularly those related to the immune system and inflammation. Its activity is tightly regulated to ensure an appropriate response to immune challenges while preventing excessive inflammation and autoimmune reactions.
NF-kappa-B is a pleiotropic transcription factor found in almost all cell types and is involved in many biological processes, including inflammation, immunity, differentiation, cell growth, tumorigenesis, and apoptosis. NF-kappa-B is a homo- or heterodimeric complex formed by the Rel-like domain-containing proteins RELA/p65, RELB, NFKB1/p105, NFKB1/p50, REL, and NFKB2/p52. These dimers bind to kappa-B sites in the DNA of their target genes. Individual dimers exhibit distinct preferences for different kappa-B sites, with varying affinities and specificities. Different dimer combinations act as transcriptional activators or repressors, respectively. NF-kappa-B is regulated by various mechanisms of post-translational modification and subcellular compartmentalization, as well as by interactions with other cofactors or corepressors.
NF-kappa-B complexes reside in the cytoplasm in an inactive state, bound to members of the NF-kappa-B inhibitor (I-kappa-B) family. In a typical activation pathway, I-kappa-B is phosphorylated by I-kappa-B kinases (IKKs) in response to various activators. This phosphorylation triggers I-kappa-B degradation, liberating the active NF-kappa-B complex, which translocates to the nucleus. NF-kappa-B heterodimeric RelB-p50 and RelB-p52 complexes are transcriptional activators. RELB does not bind to DNA or associate with RELA/p65 or REL. It stimulates promoter activity in the presence of NFKB2/p49. As a member of the NUPR1/RELB/IER3 survival pathway, it may contribute to the remarkable resistance to cell stress, such as starvation or gemcitabine treatment, observed in pancreatic ductal adenocarcinoma. It regulates the circadian clock by repressing the transcriptional activator activity of the CLOCK-ARNTL/BMAL1 heterodimer in a CRY1/CRY2-independent manner. Increased repression of the heterodimer is observed in the presence of NFKB2/p52. It is essential for both T and B lymphocyte maturation and function.
RELB is a transcription factor belonging to the NF-κB/Rel family with a molecular weight of approximately 70 kDa . It plays a critical role in the alternative (non-canonical) NF-κB signaling pathway, which regulates diverse biological processes including lymphoid organ development, B-cell maturation, and immune responses. RELB forms heterodimers with other NF-κB proteins, particularly p52, to regulate transcription of target genes.
Unlike the classical NF-κB pathway components, RELB is involved in more specialized immune functions and has gained significance in studies of lymphoid malignancies, immune disorders, and inflammatory conditions. The accurate detection and quantification of RELB is crucial for understanding its role in normal and pathological conditions, making anti-RELB antibodies indispensable research tools.
RELB monoclonal antibodies can be utilized in multiple research applications with specific protocols optimized for each technique:
For Western blotting applications, RELB is typically detected as a band at approximately 70 kDa . For optimal results in immunohistochemistry, overnight incubation at 4°C is recommended with appropriate counterstaining using hematoxylin .
RELB antibodies have been validated on several biological samples:
Human lymphoma tissue sections, where RELB localizes primarily to the cytoplasm of epithelial cells
Burkitt's lymphoma cell lines including Daudi and Raji, which express detectable levels of RELB protein
Various human, mouse, rat, and monkey samples as indicated by the species cross-reactivity data
When working with new sample types, preliminary validation experiments are essential to confirm antibody specificity and optimize detection conditions.
To maintain antibody integrity and performance, follow these guidelines:
Use a manual defrost freezer and avoid repeated freeze-thaw cycles which can compromise antibody function
For long-term storage: Store at -20°C to -70°C (up to 12 months from date of receipt)
For short-term storage: After reconstitution, store at 2-8°C under sterile conditions for up to one month
For extended storage after reconstitution: Store at -20°C to -70°C under sterile conditions for up to 6 months
Always aliquot antibodies upon first thaw to minimize freeze-thaw cycles, and centrifuge briefly before opening vials to collect all material at the bottom of the tube.
Validation of antibody specificity is crucial for reliable research results. For RELB antibodies, implement these validation approaches:
Positive control selection: Use cell lines with known RELB expression such as Daudi or Raji human Burkitt's lymphoma cell lines, which have been demonstrated to express detectable levels of RELB .
Multiple detection methods: Confirm RELB detection using complementary techniques. For example, if your primary technique is Western blotting, validate findings using immunocytochemistry or immunoprecipitation to confirm target specificity.
siRNA knockdown or CRISPR knockout: Generate RELB-depleted samples through genetic manipulation. A specific antibody should show reduced or absent signal in knockdown/knockout samples compared to wild-type controls.
Peptide competition assay: Pre-incubate the antibody with a synthetic peptide corresponding to the immunogen. If the antibody is specific, pre-incubation should block detection in subsequent assays.
Multiple antibody approach: Use different antibodies targeting distinct epitopes of RELB. Concordant results from multiple antibodies increase confidence in specificity.
When performing Western blot validation, the RELB protein should appear as a specific band at approximately 70 kDa, as demonstrated in lysates from Daudi and Raji cell lines .
Several analytical techniques can be employed to characterize RELB using monoclonal antibodies, each offering distinct advantages:
Chromatographic methods: Reversed-phase liquid chromatography (RPLC) coupled with mass spectrometry (RPLC-MS) enables separation of antibody subdomains and detection of post-translational modifications that may affect RELB binding . This approach allows both qualitative and quantitative assessment of antibody heterogeneity.
Electrophoretic techniques: Capillary electrophoresis methods, including capillary zone electrophoresis (CZE), capillary gel electrophoresis (CGE), and capillary isoelectric focusing (cIEF), offer high resolving power for separating RELB antibodies and assessing their heterogeneity based on charge and size .
Spectroscopic approaches: Nuclear Magnetic Resonance (NMR) spectroscopy, particularly 2D NMR, provides detailed structural information about RELB antibodies at the atomic level, generating molecular fingerprints that reveal high-order structural characteristics .
Surface Plasmon Resonance (SPR): This technique allows real-time measurement of binding kinetics between RELB antibodies and their target antigen, providing crucial data on affinity, avidity, and immunoreactivity .
Multi-angle light scattering (MALS): When coupled with RP-UPLC, MALS enables characterization of intact RELB antibodies and their fragments, calculating molecular weight for each chromatographic peak to identify monomeric variants and potential degradation products .
For comprehensive characterization, a combination of these techniques is recommended to assess different aspects of RELB antibody quality and function.
Post-translational modifications (PTMs) can significantly impact RELB antibody binding through several mechanisms:
Altered epitope accessibility: PTMs such as phosphorylation, glycosylation, or ubiquitination may modify the three-dimensional structure of RELB, potentially masking or exposing epitopes recognized by specific antibodies.
Charge distribution changes: Modifications that alter the charge of RELB (e.g., phosphorylation, acetylation, deamidation) can affect its electrophoretic mobility and chromatographic behavior. Ion-exchange chromatography (IEX) is particularly useful for detecting such charge variants .
Conformational changes: PTMs can induce structural changes in RELB that may impact antibody recognition, especially for antibodies targeting conformational epitopes rather than linear sequences.
Variable detection in different cellular contexts: Different cell types or conditions may produce RELB with distinct PTM patterns, leading to variable antibody reactivity across samples. For example, the phosphorylation status of RELB often differs between resting and activated immune cells.
To address the heterogeneity arising from PTMs, analytical techniques such as reversed-phase LC-MS (RPLC-MS) can be employed to separate RELB with various modifications, including pyroglutamic acid formation, isomerization, deamidation, and oxidation . This approach enables both qualitative and quantitative assessment of RELB heterogeneity.
When investigating PTMs of RELB, consider implementing peptide mapping strategies to identify modification sites and quantify modification levels at each site. This information can guide selection of appropriate antibodies that are either sensitive or insensitive to specific modifications, depending on research objectives.
The optimal conditions for using RELB antibodies vary by application:
Sample preparation: Reducing conditions show clear detection of RELB at 70 kDa
Buffer system: Use Immunoblot Buffer Group 4 for optimal results
Secondary antibody: HRP-conjugated anti-mouse/rabbit IgG depending on primary antibody source
Detection: Enhanced chemiluminescence (ECL) for sensitive detection
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Detection system: HRP-DAB (brown) with hematoxylin counterstain (blue)
Controls: Include lymphoma tissue as positive control where RELB localizes to cytoplasm in epithelial cells
Cell fixation: Immersion fixation method
Secondary antibody: Fluorochrome-conjugated (e.g., NorthernLights 557)
Expected localization: Primarily cytoplasmic in Burkitt's lymphoma cells
Lysate preparation: Use RIPA buffer with protease and phosphatase inhibitors
Protein A/G beads: Pre-clear lysate before adding antibody
Incubation: Overnight at 4°C on a rotator
Washes: Minimum of 3-5 washes with appropriate buffer to reduce background
Each technique requires optimization based on specific experimental conditions and sample types. Preliminary titration experiments are recommended to determine optimal antibody concentrations for your specific application.
When encountering inconsistent results with RELB antibodies in Western blotting, follow this systematic troubleshooting approach:
Sample preparation issues:
Ensure complete cell lysis and protein extraction
Add fresh protease inhibitors to prevent RELB degradation
Confirm protein concentration using reliable methods (BCA or Bradford assay)
Validate sample integrity by blotting for a stable housekeeping protein
Gel electrophoresis parameters:
Use appropriate acrylamide percentage (8-10% for 70 kDa RELB)
Ensure complete transfer to membrane by staining the gel post-transfer
Verify equal loading using total protein stains (Ponceau S) or housekeeping controls
Antibody-specific factors:
Detection system optimization:
Technical considerations:
If bands appear at unexpected molecular weights, evaluate potential post-translational modifications, proteolytic processing, or isoform expression. Cross-validation with orthogonal techniques (immunoprecipitation, immunocytochemistry) can help confirm the specificity of detected bands.
When designing experiments to investigate RELB in NF-κB signaling, consider these important factors:
Pathway activation specificity:
RELB primarily functions in the non-canonical NF-κB pathway
Select appropriate stimuli that specifically activate this pathway (e.g., CD40L, BAFF, lymphotoxin-β)
Include controls for canonical pathway activation (e.g., TNF-α, IL-1β) for comparison
Temporal dynamics:
Non-canonical pathway activation is typically slower than canonical signaling
Design time-course experiments that capture both early (1-6 hours) and late (12-48 hours) events
Monitor RELB nuclear translocation kinetics, which differ from classical NF-κB components
Protein interaction analysis:
Subcellular localization:
Functional readouts:
Assess transcriptional activity of RELB-dependent genes
Measure phenotypic outcomes relevant to the cell type and context
Consider pathway crosstalk with other signaling systems
Model system selection:
When interpreting results, remember that RELB functions distinctly from other NF-κB family members and exhibits cell type-specific effects. Parallel assessment of multiple NF-κB components provides a more comprehensive understanding of pathway dynamics.
Accurate quantitative assessment of RELB expression requires rigorous methodological approaches:
Western blot quantification:
Use gradient gels (4-15%) for optimal separation of the 70 kDa RELB protein
Include recombinant RELB standards at known concentrations for standard curve generation
Capture images using a digital imaging system with linear detection range
Analyze band intensity using software (ImageJ, Image Lab) with background subtraction
Normalize RELB signal to housekeeping proteins or total protein stains
Run technical triplicates to calculate coefficient of variation (CV < 15% is acceptable)
Enzyme-linked immunosorbent assay (ELISA):
Develop a sandwich ELISA using capture and detection antibodies against different RELB epitopes
Establish a standard curve using recombinant RELB protein
Validate assay parameters (sensitivity, specificity, precision, accuracy)
Calculate intra-assay and inter-assay variability to ensure reproducibility
Determine the linear range of detection for your sample types
Mass spectrometry-based quantification:
Implement targeted proteomics approaches such as selected reaction monitoring (SRM)
Use stable isotope-labeled peptide standards for absolute quantification
Select proteotypic peptides specific to RELB
Perform statistical validation of quantitative results
Flow cytometry:
Optimize cell fixation and permeabilization for intracellular RELB detection
Include isotype controls and fluorescence-minus-one (FMO) controls
Calibrate using beads with known antibody binding capacity
Report results as molecules of equivalent soluble fluorochrome (MESF)
Consider phospho-specific antibodies for activation state assessment
Real-time quantitative PCR (RT-qPCR):
Design primers specific to RELB mRNA
Validate primer efficiency (90-110%) and specificity
Use multiple reference genes for normalization
Apply the 2^(-ΔΔCt) method for relative quantification
Correlate mRNA levels with protein expression to assess post-transcriptional regulation
When comparing RELB levels across experimental conditions, ensure consistent sample preparation, equal loading, and appropriate statistical analysis to detect significant differences.
Understanding the differences between recombinant and conventional monoclonal antibodies is crucial for selecting the appropriate tool for RELB research:
Enhanced reproducibility: Recombinant antibodies are produced from sequenced DNA in defined expression systems, eliminating batch-to-batch variability inherent in hybridoma-derived antibodies .
Improved specificity: The defined sequence and production process allow for precise epitope targeting and reduced cross-reactivity with similar proteins.
Engineered modifications: Recombinant technology enables specific modifications such as humanization, isotype switching, or affinity maturation to enhance antibody performance.
Ethical considerations: Production doesn't require animals after initial sequence determination, addressing ethical concerns related to hybridoma generation and maintenance.
Consistency in post-translational modifications: When produced in controlled expression systems, recombinant antibodies show more consistent glycosylation patterns and other PTMs .
Higher production costs: Recombinant production systems typically require more sophisticated infrastructure and expertise, increasing costs.
Potential epitope constraints: Some recombinant antibodies may recognize linear epitopes more efficiently than conformational epitopes, depending on design strategy.
Expression system limitations: The choice of expression system can introduce non-native modifications that might affect antibody function.
Technical expertise requirements: Working with recombinant antibodies often requires specialized knowledge in molecular biology and protein engineering.
Feature | Recombinant Monoclonal | Conventional Monoclonal | Impact on RELB Research |
---|---|---|---|
Batch-to-batch consistency | High | Variable | Critical for longitudinal studies tracking RELB expression |
Production scalability | Unlimited once sequence established | Limited by hybridoma stability | Important for large-scale studies |
Epitope definition | Precisely defined | May vary between batches | Affects ability to detect specific RELB domains |
Post-translational modifications | Controlled | Dependent on hybridoma metabolism | May affect detection of modified RELB forms |
Customization potential | High | Limited | Enables tailored approaches for different RELB research applications |
Validation requirements | Sequence and functional validation | Functional validation only | Impacts reliability of RELB detection |
When implementing RELB antibodies in research, several critical factors must be considered to ensure reliable and reproducible results:
Experimental design should include appropriate positive controls (such as Daudi or Raji cell lysates) and negative controls to validate antibody specificity .
Optimization of protocols for specific applications is essential, with particular attention to antibody dilution, incubation conditions, and detection methods .
Understanding the molecular characteristics of RELB, including its molecular weight (70 kDa), cellular localization (primarily cytoplasmic in many cell types), and participation in the non-canonical NF-κB pathway, is fundamental to correct interpretation of results .
Comprehensive validation strategies, including multiple detection methods and analytical techniques, provide stronger evidence for observed RELB expression patterns and functional roles.
Consideration of post-translational modifications and their impact on RELB detection can help explain variability across different experimental conditions or sample types .