Recombinant TNFRSF17 Human, His is produced in two primary systems:
Binds BAFF (TNFSF13B) and APRIL (TNFSF13), activating NF-κB, MAPK8/JNK, and AKT pathways to promote plasma cell survival
Upregulates anti-apoptotic proteins (Bcl-2, Mcl-1) in multiple myeloma (MM) cells
Recombinant human TNFRSF17 protein has been produced in E. coli. It is a single, non-glycosylated polypeptide chain consisting of 130 amino acids (residues 78-184). The protein has a molecular mass of 14.1 kDa. For purification purposes, a 23 amino acid His-tag is fused to the N-terminus of TNFRSF17. The protein is purified using proprietary chromatographic techniques.
TNFRSF17 is a type I attached membrane protein of 184 amino acids that belongs to the tumor necrosis factor receptor superfamily. It is also referred to as B cell maturation antigen (BCMA) or CD269 in scientific publications. The protein possesses a conserved motif of six cysteines in the N-terminal region, forming a characteristic cysteine repeat motif that is typical of the extracellular domain of TNF receptors. This structural feature is critical for its functional interactions with ligands and subsequent downstream signaling . TNFRSF17 plays crucial roles in B cell biology, particularly in maintaining long-lived plasma cells and peripheral B cell populations.
TNFRSF17 exists as a homodimer in its native state, a structural characteristic that influences its biological activity . The protein contains the conserved cysteine-rich domains (CRDs) typical of TNF receptor family members, with the cysteine repeat motif in the extracellular domain being particularly important for ligand recognition. The protein's N-terminal region contains structural elements that facilitate receptor oligomerization, which is critical for signal transduction. Similar to other TNF receptor family members, TNFRSF17 likely contains a preligand binding assembly domain (PLAD) that facilitates receptor pre-assembly prior to ligand binding, though specific studies on TNFRSF17's PLAD are not as extensive as for other family members like CD95 and TNFR1 .
TNFRSF17 exhibits a highly specific expression pattern primarily restricted to B-lineage cells. It is expressed on both resting and activated B cells, malignant B lymphocytes, and plasma cells . This restricted expression pattern makes TNFRSF17 particularly valuable as a target for studying B cell biology and related pathologies. When designing experiments involving TNFRSF17, researchers should consider this expression profile to select appropriate cell models. B cell lines or primary B cells isolated from peripheral blood or lymphoid tissues would be suitable experimental systems, while non-B cell lineages would serve as negative controls for expression studies.
TNFRSF17 plays a crucial role in maintaining the peripheral B cell population and ensuring the survival of long-lived bone marrow plasma cells . It functions as a receptor for BAFF (TNFSF13B) and APRIL (TNFSF13), two cytokines essential for B cell development and homeostasis. Upon ligand binding, TNFRSF17 initiates signaling cascades that promote B cell survival, differentiation, and antibody production. Interestingly, soluble BCMA can act as a decoy receptor for APRIL, potentially regulating the availability of this ligand for membrane-bound receptors . TNFRSF17 mRNA expression is stimulated by IL-4 and IL-6 in murine B cells, suggesting its regulation by cytokine networks involved in humoral immunity .
TNFRSF17 binds to two main ligands: APRIL (TNFSF13) and BAFF (TNFSF13B). Notably, the interaction between TNFRSF17 and APRIL demonstrates higher affinity compared to the TNFRSF17-BAFF interaction . This differential binding affinity has important implications for downstream signaling and biological outcomes. To experimentally determine binding affinities, researchers can employ surface plasmon resonance (SPR) using purified recombinant proteins. For cellular contexts, competitive binding assays with labeled ligands can quantify relative affinities. When investigating these interactions, researchers should account for potential effects of the His-tag on binding kinetics by including appropriate controls or by enzymatic tag removal prior to binding studies.
Overexpression of TNFRSF17 in 293 cells activates multiple signaling pathways, including NF-kappa B, Elk-1, the c-Jun N-terminal kinase (JNK), and the p38 mitogen-activated protein kinase . These activations occur following TNFRSF17's association with TNF Receptor-Associated Factor (TRAF) 1, TRAF2, and TRAF3 . In breast cancer models, APRIL and BAFF-mediated activation of TNFRSF17 stimulates epithelial to mesenchymal transition and migration through the JNK signaling pathway . For experimental assessment of these pathways, researchers can utilize phospho-specific antibodies in Western blotting, luciferase reporter assays for transcription factor activation, and pharmacological inhibitors to confirm pathway specificity. RNA interference targeting specific TRAFs can further elucidate the contribution of individual adaptor proteins to TNFRSF17 signaling.
Receptor oligomerization is a critical determinant of signaling capacity for TNF receptor family members, including TNFRSF17. While the search results don't specifically address TNFRSF17 oligomerization in detail, Table 2 indicates that related receptors like TACI (which shares ligands with TNFRSF17) form at least trimeric structures, as demonstrated through SDS-PAGE of immunoprecipitates, cross-linking studies, and FRET analysis . By analogy, TNFRSF17 likely forms similar oligomeric structures that facilitate signal transduction. The oligomerization state may affect which downstream adaptor proteins are recruited and which signaling pathways are activated. To study TNFRSF17 oligomerization, researchers could employ chemical cross-linking followed by SDS-PAGE, FRET analysis with fluorescently-tagged receptor constructs, or co-immunoprecipitation studies.
To distinguish between APRIL and BAFF-mediated TNFRSF17 signaling, researchers can implement several complementary approaches. First, selective neutralizing antibodies against either APRIL or BAFF can be used to block specific ligand interactions. Second, recombinant APRIL or BAFF proteins with point mutations that disrupt binding to TNFRSF17 can serve as negative controls. Third, competitive binding assays with soluble receptor domains can determine which ligand preferentially activates TNFRSF17 in different cellular contexts. Additionally, downstream signaling kinetics and pathway activation patterns may differ between the two ligands due to their different binding affinities, which can be monitored through time-course experiments analyzing phosphorylation of signaling proteins. Gene expression profiling following stimulation with either ligand can also reveal differential transcriptional responses.
Multiple myeloma patients show elevated serum BCMA (TNFRSF17) levels, which correlate with the proportion of plasma cells in bone marrow biopsies and clinical status . This makes serum BCMA levels a potential novel biomarker for predicting outcomes in multiple myeloma patients. Researchers investigating TNFRSF17 in multiple myeloma should consider both membrane-bound and soluble forms of the protein, as they may have distinct biological roles. For experimental approaches, ELISA assays can quantify soluble BCMA in patient serum, while flow cytometry can assess membrane expression on plasma cells. Correlation analyses between BCMA levels and clinical parameters (e.g., disease stage, treatment response) can provide insights into its utility as a biomarker. In vitro studies using multiple myeloma cell lines with manipulated TNFRSF17 expression can elucidate its role in plasma cell survival and proliferation.
TNFRSF17 expression is upregulated on peripheral blood mononuclear cells (PBMCs) from mice that develop Sjogren's-like syndrome, as well as in patients with Sjogren's syndrome, rheumatoid arthritis, and on B cells from systemic lupus erythematosus (SLE) patients . This upregulation suggests a potential role for TNFRSF17 in autoimmune pathogenesis, possibly through enhanced B cell survival and autoantibody production. When studying TNFRSF17 in autoimmune contexts, researchers should consider:
Comparing expression levels between healthy donors and patients with various autoimmune conditions
Analyzing correlations between TNFRSF17 expression and disease activity scores
Examining the effects of standard immunosuppressive therapies on TNFRSF17 expression
Testing whether blockade of TNFRSF17 or its ligands ameliorates disease in animal models
Flow cytometry with cell surface staining is ideal for assessing TNFRSF17 expression on specific immune cell subsets in these disorders.
Studies have shown that both APRIL and BAFF increase epithelial to mesenchymal transition (EMT) and migratory capacity of breast cancer cells through TNFRSF17 and the JNK signaling pathway . Additionally, these ligands increase cancer stem cell numbers by enhancing the expression of pluripotency genes such as ALDH1A1, KLF4, and NANOG . To study TNFRSF17's role in breast cancer progression, researchers should employ:
Cell migration assays (wound healing, Boyden chamber) following TNFRSF17 stimulation or knockdown
EMT marker analysis (E-cadherin, vimentin, Snail, Twist) by immunoblotting and qRT-PCR
Stemness assessment using the ALDEFLUOR assay, which measures ALDH activity in live cells by flow cytometry
Sphere formation assays to evaluate self-renewal capacity
Xenograft models with TNFRSF17-manipulated breast cancer cells to assess tumor-initiating potential
Co-expression analysis of APRIL, BAFF, BCMA (TNFRSF17), and pluripotency genes using publicly available datasets, such as MetaBRIC, can provide clinical correlations .
The GEO archive contains datasets like GDS3116 that provide paired transcriptome data from letrozole (aromatase inhibitor) treated patients before and after therapy, along with their response status . Using such resources, researchers can analyze TNFRSF17 expression changes in relation to therapy response. Methodological approaches include:
Comparing pre- and post-treatment TNFRSF17 expression levels in responders versus non-responders
Correlating TNFRSF17 expression with changes in estrogen-responsive genes
Developing predictive models incorporating TNFRSF17 expression for therapy response
Using transcription factor analysis tools like ISMARA to identify regulatory factors controlling TNFRSF17 expression during treatment
For experimental validation, patient-derived xenografts or organoids treated with hormone therapy can be assessed for TNFRSF17 expression changes and correlated with growth inhibition.
When working with recombinant TNFRSF17-Fc chimera proteins, researchers should consider several factors to ensure optimal experimental outcomes. The protein should be stored according to manufacturer recommendations, typically at -80°C for long-term storage with aliquoting to avoid freeze-thaw cycles. Working solutions can be prepared at concentrations of 100-500 ng/ml in appropriate assay buffers, similar to the 100 ng/ml concentration used in APRIL and BAFF treatment experiments . Functional validation should include binding assays with known ligands (APRIL and BAFF) using techniques such as ELISA or surface plasmon resonance. When designing experiments, include appropriate controls such as isotype-matched Fc chimera proteins and consider the potential effects of the Fc portion on cellular interactions or receptor clustering.
To evaluate TNFRSF17's impact on stem cell properties, particularly in cancer models, researchers can employ the ALDEFLUOR assay as described in the literature. This method involves incubating cells (e.g., 6 million cells/ml) with the ALDEFLUOR reagent, a fluorescent substrate for aldehyde dehydrogenase (ALDH), for approximately 45 minutes at 37°C . Control samples should include the specific ALDH inhibitor dimethylamino benzaldehyde (DEAB). Flow cytometry can then be used to measure the fluorescent reaction product, which is proportional to ALDH activity, using identical instrument settings for test and control samples . Additionally, researchers should assess expression of pluripotency genes such as ALDH1A1, KLF4, and NANOG using qRT-PCR or immunoblotting following TNFRSF17 stimulation or inhibition . Sphere formation assays in low-attachment conditions can provide functional validation of stemness properties.
TNF receptors are categorized based on their response to soluble ligand trimers. Category I TNFRs (like TNFR1 and LTβR) are robustly activated by soluble ligand trimers, while Category II TNFRs (including 4-1BB, CD27, CD40, CD95, OX40, TNFR2) require membrane-bound or aggregated ligands for efficient signaling . To determine TNFRSF17's classification, researchers should:
Compare cellular responses to soluble versus membrane-bound ligands
Assess activation of downstream signaling pathways (NF-κB, JNK) after stimulation with different ligand forms
Evaluate receptor clustering using microscopy techniques following exposure to soluble and membrane-bound ligands
Examine the effects of artificially clustered soluble ligands (e.g., using antibody cross-linking)
Based on the literature, TNFRSF17 (BCMA) appears similar to other TNFRs that efficiently interact with soluble BAFF trimers but may require higher-order clustering for optimal signaling .
Pre-ligand assembly domains (PLADs) have been identified in several TNF receptor family members, primarily in the N-terminal CRD1 . To study potential PLAD-mediated assembly of TNFRSF17, researchers can employ several complementary approaches:
Size exclusion chromatography to analyze the oligomeric state of the recombinant extracellular domain
Cross-linking studies using chemical cross-linkers followed by SDS-PAGE analysis
FRET analysis with fluorescently tagged receptor constructs
Co-immunoprecipitation of differently tagged TNFRSF17 variants
Mutagenesis of potential PLAD residues in the N-terminal region
Additionally, researchers could generate dimeric fusion proteins of the putative PLAD domain with glutathione S-transferase or Fc fragments, similar to approaches used with TNFR1, to test their ability to inhibit TNFRSF17 signaling, which would suggest competition for pre-ligand assembly .
TNFRSF17 receptor oligomerization can be compared to other TNF receptor family members using the data in Table 2 from the search results . While specific information about TNFRSF17 oligomerization is not directly provided, related receptors show various oligomerization states and mechanisms. For instance, some receptors like CD27, P75NGFR, and fractions of CD40 and 41BB form dimers through disulfide linkages . Others like TACI (which shares ligands with TNFRSF17) form at least trimers, as demonstrated through various techniques including SDS-PAGE of immunoprecipitates, cross-linking, and FRET . To experimentally compare TNFRSF17 oligomerization with other family members, researchers should employ multiple complementary techniques, including those mentioned in the table: SDS-PAGE analysis of immunoprecipitated receptors, cross-linking studies, FRET analysis, and size exclusion chromatography. This comparative approach would provide insights into whether TNFRSF17 follows patterns similar to other BAFF/APRIL receptors or has unique oligomerization properties.
When studying TNFRSF17 in different cellular systems, researchers must consider several methodological factors to ensure robust and reproducible results:
Expression level assessment: Quantify endogenous TNFRSF17 expression in the cellular system of interest using flow cytometry, Western blotting, or qRT-PCR before initiating functional studies.
Ligand selection: Consider the differential binding affinities of APRIL versus BAFF when designing stimulation experiments .
Signal pathway analysis: Include both early (minutes to hours) and late (hours to days) timepoints when assessing signaling responses, as TNFRSF17 activation affects both immediate signaling cascades (e.g., JNK, p38, NF-κB) and longer-term gene expression changes .
Cellular context: Account for the expression of other BAFF/APRIL receptors (TACI, BAFFR) that might influence ligand availability and signaling outcomes.
Experimental controls: Include receptor-blocking antibodies or soluble receptor domains as negative controls to confirm the specificity of observed effects.
For heterologous expression systems, consider using inducible expression systems to control TNFRSF17 levels and avoid potential artifacts from continuous overexpression.
To investigate therapeutic targeting of the TNFRSF17 pathway, researchers can employ several strategic approaches:
Blocking strategies:
Develop neutralizing antibodies against the extracellular domain of TNFRSF17
Design soluble decoy receptors based on the TNFRSF17 extracellular domain to sequester ligands
Create small molecule inhibitors targeting the TNFRSF17-ligand interaction interface
Develop ligand traps using fusion proteins
Signaling inhibition:
Preclinical testing:
Establish relevant disease models where TNFRSF17 plays a documented role
For multiple myeloma, use patient-derived xenografts that maintain TNFRSF17 expression
For autoimmune models, consider transgenic mice with altered TNFRSF17 expression
Biomarker development:
Correlate serum soluble TNFRSF17 levels with disease progression and treatment response
Develop companion diagnostics to identify patients most likely to benefit from TNFRSF17-targeted therapies
The success of therapeutic approaches targeting other TNF receptor family members provides precedent for this strategy.
Computational approaches to predict TNFRSF17 involvement in gene regulatory networks can utilize both publicly available datasets and novel analysis methods:
Co-expression analysis: Analyze co-expression patterns between TNFRSF17, its ligands, and other genes across large datasets, as demonstrated in the MetaBRIC study of 2509 samples, which examined co-expression of APRIL, BAFF, BCMA, androgen receptor, and pluripotency genes .
Transcription factor analysis: Employ web resources like ISMARA to predict transcription factor modifications through gene transcript changes, as was done in the analysis of letrozole-treated patient data .
Network inference algorithms: Apply algorithms such as WGCNA (Weighted Gene Co-expression Network Analysis) to identify gene modules associated with TNFRSF17 expression across different conditions.
Pathway enrichment analysis: Conduct Gene Ontology and pathway enrichment analyses on genes correlated with TNFRSF17 to identify biological processes potentially regulated by this receptor.
Machine learning approaches: Develop predictive models that can identify potential regulatory relationships between TNFRSF17 and other genes based on expression patterns across multiple datasets.
B-Cell Maturation Antigen (BCMA), also known as tumor necrosis factor receptor superfamily member 17 (TNFRSF17), is a protein encoded by the TNFRSF17 gene in humans . BCMA is a cell surface receptor that plays a crucial role in the regulation of B cell proliferation, survival, and maturation into plasma cells . The recombinant form of BCMA, tagged with a His-tag, is widely used in research and therapeutic applications.
The human recombinant BCMA with a His-tag is typically produced in various expression systems such as E. coli or baculovirus . The His-tag, a sequence of histidine residues, is added to the protein to facilitate purification using immobilized metal affinity chromatography (IMAC). This tag does not interfere with the protein’s function and allows for efficient isolation of the recombinant protein.
For example, TNFRSF17 Human Recombinant produced in E. coli is a single, non-glycosylated polypeptide chain containing 130 amino acids and a 23 amino acid His-tag at the N-terminus . Another variant produced in baculovirus is a glycosylated polypeptide chain containing 296 amino acids and a 242 amino acid hIgG-His-Tag at the C-terminus .
BCMA is primarily expressed on mature B lymphocytes and plasma cells . It is a receptor for B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL), both of which are crucial for B cell development and immune response . The interaction of BCMA with these ligands leads to the activation of NF-kappaB and MAPK8/JNK pathways, promoting cell survival and proliferation .
The expression and function of BCMA are regulated by several mechanisms, including transcriptional and post-transcriptional regulation . The binding of BAFF and APRIL to BCMA triggers signaling pathways that regulate B cell survival and differentiation . Additionally, soluble BCMA (sBCMA), a cleaved form of the receptor, can be found in the serum and is often elevated in patients with multiple myeloma .
BCMA has emerged as a promising therapeutic target, particularly in the treatment of multiple myeloma . Anti-BCMA therapies, including monoclonal antibodies, antibody-drug conjugates, and chimeric antigen receptor (CAR) T-cell therapies, have shown significant efficacy in clinical trials . These therapies target BCMA-expressing cells, leading to their destruction and providing a potential treatment for refractory or relapsed multiple myeloma .