TNFRSF17, also known as B-cell maturation antigen (BCMA) or CD269, is a type III transmembrane protein belonging to the TNF receptor superfamily. The partial recombinant form typically includes the extracellular domain (aa 1–54 or 78–184) fused with affinity tags (e.g., His, Myc, Fc) for purification and detection. This truncated variant retains ligand-binding capability critical for studying B-cell signaling and therapeutic targeting .
The recombinant protein is produced via bacterial (e.g., E. coli) or mammalian (e.g., HEK293) expression systems, with purification optimized for low endotoxin levels (<1 EU/μg) . Bioactivity is confirmed through ligand-binding assays and functional studies:
TNFRSF17 is a pivotal target in B-cell malignancies, particularly multiple myeloma (MM). Its partial recombinant form facilitates drug development and diagnostic research:
Approach | Mechanism | Example |
---|---|---|
CAR-T Cell Therapy | Guides T cells to target BCMA+ MM cells; transient expression limits toxicity | Descartes-08 mRNA CAR-T |
Antibody-Drug Conjugates | Deliver cytotoxic agents to BCMA+ cells | Belantamab mafodotin |
Bispecific Antibodies | Engage T cells and BCMA+ MM cells | Teclistamab |
Recent studies highlight TNFRSF17’s role in oncology and immune regulation:
Objective: Evaluate anti-BCMA CAR-T cells in MM.
Method: TNFRSF17-coated beads were used to assess CAR-T activation and cytotoxicity.
Result: CAR-T cells showed dose-dependent cytolysis correlating with BCMA expression duration .
Objective: Investigate TNFRSF17’s role in 25-HC-induced metastasis.
Method: ERβ knockdown reduced TNFRSF17 expression, inhibiting proliferation/migration.
Result: TNFRSF17 knockdown replicated these effects, implicating it in metastatic pathways .
TNFRSF17 interacts with ligands and signaling adaptors to regulate B-cell survival:
Interaction Partner | Role | Pathway Activation |
---|---|---|
BAFF/APRIL | Ligands binding to TNFRSF17’s extracellular domain | NF-κB, JNK/MAPK8, AKT |
TRAF5/6 | Adapter proteins recruiting downstream kinases | NF-κB, MEK/ERK, p38 |
Form | Advantages | Limitations |
---|---|---|
His-tagged (E. coli) | High purity, low cost | Non-glycosylated; reduced affinity |
Fc-tagged (HEK293) | Glycosylated; mimics native binding | Higher production costs |
R-PE Labeled | Enables FACS-based detection of BCMA+ cells | Limited to diagnostic applications |
This Recombinant Human TNFRSF17 protein is intended for cancer research. TNFRSF17, also known as Tumor necrosis factor receptor superfamily member 17 or B-cell maturation protein (BCMA), plays a crucial role in B-cell development and is implicated in various cancer-related processes.
Our protein is produced using an E. coli expression system and encompasses amino acids 5 to 54, representing a portion of the TNFRSF17 sequence. It is tag-free, ensuring its native conformation and eliminating potential interference in downstream applications. With a purity exceeding 98% and minimal endotoxin contamination, our TNFRSF17 protein guarantees reliable and consistent results.
The protein's activity has been validated. The lyophilized powder form ensures easy handling and storage, providing convenience for your research endeavors. Investigate the potential of TNFRSF17 in cancer biology and unravel its role in B-cell maturation and tumorigenesis with our high-quality and bioactive protein.
This receptor for TNFSF13B/BLyS/BAFF and TNFSF13/APRIL promotes B-cell survival and plays a role in regulating humoral immunity. It activates NF-kappa-B and JNK pathways.
TNFRSF17, also known as B-cell maturation antigen (BCMA), BCM, CD269, or TNFRSF13A, is a cell surface receptor belonging to the tumor necrosis factor receptor superfamily. It is encoded by the TNFRSF17 gene in humans and primarily expressed on mature B lymphocytes . The protein functions as a receptor that recognizes B-cell activating factor (BAFF) and plays significant roles in B-cell development, autoimmune responses, and various pathological conditions . In scientific research, it's important to search literature using all variant names to ensure comprehensive coverage of relevant publications.
The recombinant partial TNFRSF17 protein typically contains the extracellular domain (such as amino acids 1-54) fused with an hFc-tag at the C-terminal . This differs from the full-length native protein which includes the transmembrane and intracellular domains necessary for signal transduction. The partial recombinant protein maintains the binding domain (BCMA TALL-1 binding domain) required for interaction with its ligand TNFSF13B , but lacks the ability to independently initiate downstream signaling pathways like NF-kappaB and MAPK8/JNK. The molecular weight of the partial recombinant protein (approximately 34.8 kDa) differs from the native protein due to the truncation and addition of tags designed to facilitate purification and detection in experimental settings .
The key functional domain of TNFRSF17 is the BCMA TALL-1 binding domain located at the N-terminus, which is required for binding to TNFSF13B (BAFF) . This conserved domain is crucial for the protein's biological activity. When designing experiments involving TNFRSF17, researchers must ensure that this domain remains intact and properly folded in recombinant constructs.
For experimental design, considerations should include:
Verifying binding capability through functional assays (ELISA, LSPR) with known ligands
Ensuring proper protein folding by quality control methods
Using appropriate buffer conditions to maintain structural integrity
Determining whether the experimental question requires the full signaling capability (needing full-length protein) or just binding interactions (where partial protein may suffice)
Including appropriate controls to distinguish effects of the protein from those of any fusion tags
Recombinant TNFRSF17 serves as an excellent standard in ELISA-based detection methods due to its high purity and well-characterized binding properties. To implement TNFRSF17 as an ELISA standard:
Coat ELISA plates with purified recombinant TNFRSF17 at varying concentrations (typically 0.1-10 μg/ml) to establish a standard curve
Block non-specific binding sites with appropriate blocking buffer
Add the ligand (such as biotinylated TNFSF13B) followed by detection reagents
Analyze binding curves to determine EC50 values (typically in the range of 0.31-0.57 ng/ml for high-quality preparations)
For quantitative detection of TNFRSF17 in patient samples, the recombinant protein provides a reliable reference point for calibration. When developing such assays, researchers should verify batch consistency through quality control measures including SDS-PAGE analysis for purity (>90% recommended) and endotoxin testing (<1.0 EU/μg using LAL method) to ensure reliable and reproducible results .
The optimal experimental conditions for studying TNFRSF17 interactions with its ligands (particularly TNFSF13B/BAFF) include:
Buffer composition:
Physiological pH (7.2-7.4)
PBS or TBS with 0.05-0.1% non-ionic detergent for reduced non-specific binding
1-5% BSA or similar blocking protein
Calcium (1-2 mM) to maintain protein stability
Analytical techniques:
Surface Plasmon Resonance (SPR) or LSPR for real-time binding kinetics
ELISA for endpoint measurements
Flow cytometry for cell-surface expression studies
Co-immunoprecipitation for complex formation analysis
Important controls:
Negative control using non-relevant proteins of similar size
Competitive binding assays using known ligands
Validation with multiple techniques to confirm specificity
Researchers should maintain sample integrity by avoiding freeze-thaw cycles and using freshly prepared reagents when possible. For kinetic studies, temperature control (typically 25°C or 37°C) is essential for reproducible results .
To effectively use recombinant TNFRSF17 in cell-based assays studying B-cell function:
Cell selection: Primary B cells, plasma cells, or B-cell lines (such as those derived from multiple myeloma) that express the complementary ligands
Assay formats:
Stimulation assays: Use plate-bound or soluble recombinant TNFRSF17 to stimulate cells expressing TNFSF13B
Inhibition assays: Use recombinant TNFRSF17 to competitively inhibit TNFSF13B-TNFRSF17 interactions
Cell viability/proliferation assays: Measure effects on B-cell survival using MTT, XTT, or flow cytometry
Functional readouts:
NF-κB activation (using reporter cell lines or Western blotting)
MAPK8/JNK pathway activation
Changes in gene expression profiles
B-cell survival and proliferation metrics
Antibody production measurement
Optimization strategies:
Titrate protein concentrations (typically 1-100 ng/ml)
Determine optimal time points for measuring different responses
Include pathway inhibitors as controls to confirm specificity
When designing these experiments, researchers should consider the partial nature of recombinant TNFRSF17 proteins which may affect certain signaling outcomes compared to the native receptor .
TNFRSF17 expression is significantly upregulated in multiple myeloma (MM) compared to normal plasma cells. Specifically:
Expression patterns:
Therapeutic implications:
Resistance mechanisms:
Biallelic deletion of TNFRSF17 (TNFSF17) has been identified as a rare but important mechanism of antigen escape and resistance to BCMA-targeted therapies
Extracellular domain missense mutations (e.g., R27P) can mediate resistance to T-cell engagers
Preexisting subclonal TNFRSF17 deletion (occurring in approximately 5% of patients prior to CAR T-cell therapy) may predict treatment resistance
These findings underscore the need for monitoring TNFRSF17 structural changes and expression levels before and during treatment. Combination approaches targeting multiple epitopes or alternative B-cell markers may help overcome resistance mechanisms in clinical settings .
TNFRSF17 plays a significant role in lupus nephritis (LN) pathogenesis with strong therapeutic implications:
Expression patterns:
Clinical correlations:
Potential therapeutic approaches:
TNFRSF17-targeting drugs like IBI379 (a construct with anti-TNFRSF17 chains and anti-CD3-ScFv-Fc fusion)
These agents can effectively induce apoptosis in patient plasma cells with minimal effects on B cells
May address the extrafollicular B cell response characteristic of childhood idiopathic nephrotic syndrome
Mechanistically, TNFRSF17 mediates plasma cell survival through the classical NF-κB pathway, suggesting that targeting this receptor could disrupt pathogenic antibody production. The specific correlation with disease markers and the demonstrated efficacy of experimental TNFRSF17-targeting agents makes this receptor a promising therapeutic target for lupus nephritis .
Researchers can effectively detect and quantify TNFRSF17 deletion and mutations in patient samples using several complementary approaches:
Genomic analysis:
Single-cell approaches:
Protein detection:
Flow cytometry for surface expression quantification
Immunohistochemistry for tissue localization
ELISA for soluble/cleaved forms (sBCMA)
Mutation analysis:
For clinical relevance, researchers should monitor TNFRSF17 status before treatment initiation and at relapse, particularly in patients receiving BCMA-targeted therapies. Sensitivity is crucial—techniques should detect subclonal deletions with variant allelic fractions as low as 3.5%, as these can impact treatment response . When analyzing results, researchers should consider the size of deletions (approximately 0.8 Mb pairs encompassing the TNFRSF17 gene locus has been reported) and distinguish between monoallelic and biallelic losses .
Developing multi-epitope targeting approaches to overcome TNFRSF17 antigen escape requires systematic strategies:
Epitope mapping and selection:
Identify conserved epitopes across the TNFRSF17 protein using crystallographic data
Target multiple non-overlapping epitopes simultaneously
Include epitopes from both N-terminal and C-terminal regions
Focus on epitopes critical for ligand binding or signaling
Combination therapy design:
Develop cocktails of monoclonal antibodies targeting different TNFRSF17 epitopes
Create bispecific or multispecific antibodies that engage TNFRSF17 and additional B-cell markers
Combine TNFRSF17-targeted therapy with agents targeting alternative B-cell pathways
CAR T-cell engineering approaches:
Design CAR T-cells with multiple single-chain variable fragments recognizing different TNFRSF17 epitopes
Develop dual-targeting CAR T-cells recognizing both TNFRSF17 and secondary targets (e.g., CD38, SLAMF7)
Incorporate logic-gated CAR designs requiring recognition of multiple antigens
Monitoring and adaptation strategies:
This comprehensive approach acknowledges that biallelic deletion of TNFRSF17 and extracellular domain mutations represent important mechanisms of resistance to BCMA-targeted therapies. By simultaneously targeting multiple epitopes or combining TNFRSF17 targeting with other approaches, researchers can potentially prevent or overcome antigen escape phenomena .
To determine the functional consequences of TNFRSF17 missense mutations on binding to therapeutic antibodies, researchers should employ a systematic multi-technique approach:
Structural analysis:
Computational modeling of mutations using protein structure prediction tools
X-ray crystallography or cryo-EM of mutant-antibody complexes
Molecular dynamics simulations to predict binding energy changes
Binding kinetics assessment:
Surface plasmon resonance (SPR) comparing wild-type and mutant TNFRSF17 binding to therapeutic antibodies
Bio-layer interferometry to determine kon and koff rates
Isothermal titration calorimetry for thermodynamic binding parameters
Cell-based functional assays:
Generate cell lines expressing wild-type or mutant TNFRSF17 (e.g., R27P)
Flow cytometry to quantify antibody binding to cell surface
Antibody-dependent cellular cytotoxicity (ADCC) assays
Complement-dependent cytotoxicity (CDC) assays
Cell killing assays with antibody-drug conjugates or T-cell engagers
In vitro mutation screening:
Validation in patient-derived samples:
This comprehensive approach allows researchers to identify which mutations significantly impact therapeutic binding, understand the structural basis for resistance, and potentially design next-generation therapeutics that maintain efficacy against mutant forms of TNFRSF17 .
Recombinant TNFRSF17 offers valuable tools for studying extrafollicular B cell responses in autoimmune conditions through several methodological approaches:
Ex vivo analysis of patient samples:
Use labeled recombinant TNFRSF17 to identify cells expressing complementary ligands
Develop flow cytometry panels incorporating TNFRSF17 binding to characterize extrafollicular B cell subsets
Perform competitive binding assays with patient serum to detect autoantibodies targeting TNFRSF17-related pathways
In vitro functional studies:
Signaling pathway interrogation:
Animal model applications:
This approach is particularly relevant given recent findings that extrafollicular B cell responses, marked by expansion of atypical B cells and antibody-secreting cells, represent the major immune perturbation in conditions like childhood idiopathic nephrotic syndrome and lupus nephritis. TNFRSF17 expression has been found to correlate positively with disease markers such as urinary protein levels, suggesting its central role in these pathogenic processes .
Critical quality control parameters for validating recombinant TNFRSF17 activity include:
Purity assessment:
Endotoxin testing:
Functional validation:
Structural integrity:
Circular dichroism to verify proper protein folding
Size exclusion chromatography to detect aggregation
Thermal shift assays to assess stability
Batch consistency:
These quality control measures ensure that experimental outcomes reflect true biological activity rather than artifacts from impurities, misfolding, or degradation. For researchers conducting critical experiments, validating multiple parameters rather than relying on a single quality metric provides greater confidence in results and improves reproducibility across laboratories .
Distinguishing between the effects of full-length and partial TNFRSF17 in experimental systems requires strategic experimental design:
Complementary expression systems:
Generate matched cell lines expressing either full-length or partial TNFRSF17
Ensure equivalent expression levels through quantitative flow cytometry
Use inducible expression systems for temporal control
Domain-specific functional assays:
Binding assays: Both full-length and partial proteins should demonstrate comparable ligand binding
Signaling assays: Only full-length protein will initiate downstream signaling cascades
Monitor NF-κB and MAPK8/JNK activation using reporter systems or phosphorylation studies
Competitive experimental designs:
Use partial protein as a competitive inhibitor of full-length protein
Titrate partial protein to determine binding site saturation effects
Analyze dose-response relationships for both variants
Molecular controls:
Readout selection:
Select proximal readouts (e.g., receptor clustering) and distal readouts (e.g., gene expression)
Time-course experiments to distinguish immediate vs. delayed effects
Single-cell analyses to capture heterogeneous responses
By implementing these strategies, researchers can clearly delineate which experimental observations are attributable to the binding/competitive functions of the partial protein versus the complete signaling capabilities of the full-length receptor .
Best practices for handling and storing recombinant TNFRSF17 to maintain optimal activity include:
Storage conditions:
Store lyophilized protein at -20°C or -80°C
After reconstitution, aliquot and store at -80°C to minimize freeze-thaw cycles
Avoid storage in frost-free freezers due to temperature fluctuations
Reconstitution protocol:
Use sterile, molecular biology-grade water or buffer
For higher concentration needs, reconstitute in minimal volume and further dilute in working buffer
Allow protein to fully dissolve (typically 10-20 minutes at room temperature) with gentle swirling rather than vortexing
Working solution preparation:
Optimal buffer composition: PBS or TBS with 0.1% carrier protein (BSA or HSA)
pH range: 7.2-7.4
Optional additives: 0.05% sodium azide for extended storage, protease inhibitors for sensitive applications
Filter-sterilize working solutions (0.22 μm filter)
Stability considerations:
Minimize freeze-thaw cycles (ideally ≤3)
Maintain protein at 2-8°C during experiment preparation (not room temperature)
Discard diluted working solutions after 24 hours
Monitor solution clarity; cloudiness indicates potential aggregation
Quality verification before critical experiments:
Following these practices ensures that experimental outcomes reflect true biological activity rather than artifacts from protein degradation or denaturation. For critical applications, researchers should document handling procedures and include appropriate controls to verify protein functionality at the time of experiment .
Single-cell technologies offer powerful approaches to understand TNFRSF17 expression heterogeneity in disease states:
Single-cell RNA sequencing applications:
Identify previously unrecognized cell populations with differential TNFRSF17 expression
Map TNFRSF17 expression across B-cell differentiation trajectories
Correlate TNFRSF17 expression with broader transcriptional programs
Detect rare resistant cell populations before they expand under therapeutic pressure
Single-cell protein analysis:
Mass cytometry (CyTOF) to simultaneously measure TNFRSF17 and dozens of other proteins
Imaging mass cytometry for spatial context in tissue samples
Single-cell Western blotting for protein isoform discrimination
Genomic heterogeneity assessment:
Integrated multi-omics approaches:
CITE-seq to simultaneously measure surface TNFRSF17 protein and mRNA expression
Single-cell ATAC-seq to determine chromatin accessibility at the TNFRSF17 locus
Spatial transcriptomics to map TNFRSF17 expression in tissue context
Recent findings using these technologies have already revealed important insights, such as identifying the extrafollicular B cell response as a major immune perturbation in childhood idiopathic nephrotic syndrome, characterized by expansion of atypical B cells and antibody-secreting cells with distinctive TNFRSF17 expression patterns . Similarly, single-cell approaches have detected rare subclonal TNFRSF17 deletions in approximately 5% of multiple myeloma patients prior to CAR T-cell therapy, with significant implications for treatment response .
Emerging approaches for developing next-generation TNFRSF17-targeted therapeutics include:
Novel antibody engineering:
Advanced cellular therapies:
Innovative conjugate technologies:
Alternative modalities:
Proteolysis targeting chimeras (PROTACs) directing TNFRSF17 for degradation
mRNA-encoded TNFRSF17-targeting therapeutics
Small molecule degraders of TNFRSF17
Engineered TNFSF13B variants with enhanced binding
Predictive biomarker integration:
These approaches address key limitations of current TNFRSF17-targeted therapies, including antigen escape through TNFRSF17 deletion or mutation, limited durability of response, and on-target off-tumor toxicity. Particularly promising are combinations that can effectively induce apoptosis in malignant or pathogenic plasma cells while minimizing effects on normal B cells, as demonstrated with agents like IBI379 in lupus nephritis models .
Systems biology approaches offer powerful frameworks for integrating TNFRSF17 signaling into broader B cell regulatory networks:
Network modeling strategies:
Construct protein-protein interaction networks centered on TNFRSF17
Develop dynamic mathematical models of TNFRSF17 signaling pathways (NF-κB and MAPK8/JNK)
Integrate transcriptional regulatory networks downstream of TNFRSF17 activation
Create multi-scale models connecting molecular events to cellular behaviors
Multi-omics data integration:
Comparative network analysis:
Compare TNFRSF17 signaling networks across:
Normal vs. malignant B cells
Different B cell developmental stages
Various autoimmune conditions
Treatment-responsive vs. resistant states
Perturbation biology approaches:
This systems approach has already yielded important insights. For instance, analysis of lupus nephritis identified TNFRSF17 as part of a signature of 70 LN-specific genes significantly enriched in the "regulation of biological quality" GO term and cell cycle pathway . Similarly, in childhood idiopathic nephrotic syndrome, systems analysis revealed that a B cell transcriptional program poised for effector functions, with TNFRSF17 as a key component, represents the major immune perturbation .
By applying these approaches, researchers can move beyond studying TNFRSF17 in isolation to understand how it functions within the complex regulatory landscape of B cells in both health and disease, potentially identifying novel therapeutic targets and biomarkers.