TNFRSF10B is a transmembrane protein with an extracellular cysteine-rich domain, a transmembrane region, and an intracellular death domain (DD) essential for apoptosis signaling . It binds TRAIL, leading to receptor oligomerization and recruitment of FADD (Fas-associated death domain), which activates caspase-8 and triggers apoptosis . Key structural features include:
Amino Acid Sequence: 394 residues (56–210 a.a.), with a C-terminal hIgG-His-Tag in recombinant forms .
Molecular Weight: ~43.9 kDa (predicted), but SDS-PAGE migration appears between 40–57 kDa due to glycosylation and tags .
Isoforms: Two transcript variants encoding distinct isoforms and one non-coding transcript .
Recombinant TNFRSF10B is expressed in Sf9 insect cells, which provide eukaryotic post-translational modifications while avoiding mammalian system complexities. Key production parameters include:
Higher Yield: Sf9 cells enable scalable production with consistent glycosylation patterns (Man3, Man3F, Man6) .
Cost Efficiency: Lower production costs compared to mammalian systems .
TNFRSF10B is a therapeutic target in cancers (e.g., squamous cell carcinoma) due to its role in TRAIL-induced apoptosis . Recombinant TNFRSF10B is used to:
Mimic TRAIL Receptor Activation: Induce apoptosis in cancer cells while sparing normal cells .
Study Resistance Mechanisms: Identify pathways that inhibit TNFRSF10B-mediated apoptosis .
Mouse TRAIL R2/Fc chimera (TNFRSF10B-Fc) neutralizes TRAIL, preventing apoptosis in experimental models . This tool is critical for studying TRAIL’s role in inflammation and immune regulation .
TNFRSF10B is essential for ER stress-induced apoptosis, as shown in knockout models .
The ExpiSf system (high-density insect cell culture) outperforms traditional Sf9 workflows in protein yield and efficiency :
Biological Activity: TNF-α produced via ExpiSf retains equivalent activity to traditional methods, as validated by NF-κB luciferase assays .
TNFRSF10B is implicated in:
Squamous Cell Carcinoma: Overexpression correlates with TRAIL sensitivity .
Laryngeal Cancer: Targeted therapies leveraging TNFRSF10B/TRAIL axis are under investigation .
Recombinant TNFRSF10B demonstrates functional activity in:
TNFRSF10B, CD262, DR5, KILLER, KILLER/DR5, TRAIL-R2, TRAILR2, TRICK2, TRICK2A, TRICK2B, TRICKB, ZTNFR9, Death receptor 5, TNF-related apoptosis-inducing ligand receptor 2.
Sf9, Insect cells.
ITQQDLAPQQ RAAPQQKRSS PSEGLCPPGH HISEDGRDCI SCKYGQDYST HWNDLLFCLR CTRCDSGEVE LSPCTTTRNT VCQCEEGTFR EEDSPEMCRK CRTGCPRGMV KVGDCTPWSD IECVHKESGT KHSGEVPAVE ETVTSSPGTP ASPCSLEPKS CDKTHTCPPC PAPELLGGPS
VFLFPPKPKD TLMISRTPEV TCVVVDVSHE DPEVKFNWYV DGVEVHNAKT KPREEQYNST YRVVSVLTVL HQDWLNGKEY KCKVSNKALP APIEKTISKA KGQPREPQVY TLPPSRDELT KNQVSLTCLV KGFYPSDIAV EWESNGQPEN NYKTTPPVLD SDGSFFLYSK LTVDKSRWQQ
GNVFSCSVMH EALHNHYTQK SLSLSPGKHH HHHH.
TNFRSF10B (also known as Death Receptor 5, DR5, CD262, KILLER, TRAIL-R2) is a member of the TNF-receptor superfamily of transmembrane proteins. It contains a cytoplasmic "death domain" capable of activating the cell's apoptotic machinery. This receptor is activated by binding to either membrane-anchored or soluble TRAIL/Apo2L, initiating a signaling cascade that leads to programmed cell death. TNFRSF10B is a critical mediator in the extrinsic apoptosis pathway, making it an important target in cancer research due to its potential to selectively induce apoptosis in tumor cells .
Sf9 refers to Spodoptera frugiperda (fall armyworm) insect cells used as an expression system for recombinant proteins. For TNFRSF10B research, the Sf9 baculovirus expression system offers several advantages: it provides proper eukaryotic post-translational modifications (particularly glycosylation), yields high expression levels, and produces proteins with structural integrity similar to mammalian-expressed proteins. The TNFRSF10B Human, Sf9 preparation specifically refers to human TNFRSF10B produced in this insect cell expression system, which is ideal for functional studies where properly folded and glycosylated protein is required .
TNFRSF10B Human, Sf9 is a single, glycosylated polypeptide chain containing 394 amino acids (positions 56-210 of the native sequence) with a molecular mass of 43.9kDa. On SDS-PAGE analysis, it typically appears at approximately 40-57kDa due to glycosylation. In the recombinant preparation, it is expressed with a 239 amino acid hIgG-His tag at the C-Terminus to facilitate purification. The preparation is purified by proprietary chromatographic techniques to greater than 90% purity as determined by SDS-PAGE .
For short-term use (2-4 weeks), TNFRSF10B protein solution can be stored at 4°C. For longer storage periods, it should be kept frozen at -20°C. The standard formulation contains Phosphate Buffered Saline (pH 7.4) and 10% glycerol. For long-term storage, it is recommended to add a carrier protein (0.1% HSA or BSA) to enhance stability. Multiple freeze-thaw cycles should be avoided as they can lead to protein denaturation and loss of activity. Aliquoting the protein before freezing is advised for research requiring multiple experimental timepoints .
The functional activity of TNFRSF10B Human, Sf9 can be measured by its ability to inhibit cytotoxicity in assays using Jurkat human T lymphocyte cells. The ED50 (effective dose for 50% inhibition) for this effect is typically ≤5ng/ml with TRAIL. This cytotoxicity inhibition assay serves as the standard functional assessment for TNFRSF10B activity. Alternative methods include binding assays with labeled TRAIL, apoptosis assays in susceptible cell lines, and co-immunoprecipitation studies to assess interaction with downstream signaling components .
When designing experiments with TNFRSF10B Human, Sf9, researchers should include:
Positive controls: Cells known to express functional TRAIL receptors (e.g., Jurkat cells)
Negative controls: Cell lines with low/no TNFRSF10B expression or TNFRSF10B-knockout cells
Dose-response curves: Serial dilutions of TNFRSF10B to establish proper working concentrations
Specificity controls: Other TNF receptor family members to confirm specificity of observed effects
Vehicle controls: Buffer-only treatments to account for any effects from the storage solution
These controls help ensure experimental validity and assist in troubleshooting unexpected results when working with this protein .
Table 1: TNFRSF10B Polymorphisms Associated with NSCLC Survival
SNP ID | Risk Allele | Hazard Ratio | 95% Confidence Interval |
---|---|---|---|
rs11785599 | T | 1.41 | 1.16-1.70 |
rs1047275 | C | 1.35 | 1.11-1.65 |
rs4460370 | T | 1.29 | 1.06-1.57 |
rs883429 | T | 1.24 | 0.99-1.54 |
TNFRSF10B/DR5 expression is inducible by cancer therapeutic agents, including platinum agents and taxanes commonly used as adjuvant chemotherapy for lung cancer patients. Upregulation of TNFRSF10B/DR5 can induce programmed cell death or augmentation of TRAIL-induced apoptosis. Research has shown that TNFRSF10B polymorphisms had treatment-specific effects: risk-allele haplotypes exhibited a statistically significant increased risk of death among patients who underwent surgery only, but no significant effects among patients who received surgery and adjuvant chemotherapy. This suggests that TNFRSF10B polymorphisms might serve as predictive biomarkers for determining which patients would benefit from adjuvant chemotherapy following surgical resection .
Several methodologies are employed to study TNFRSF10B-mediated apoptosis in tumor models:
Apoptosis assays: Annexin V/PI staining followed by flow cytometry to quantify early and late apoptotic cells
Caspase activity measurements: Fluorometric or colorimetric assays for caspase-8, caspase-10, and caspase-3 activation
Death receptor expression analysis: Immunoblotting, immunohistochemistry, and flow cytometry to assess TNFRSF10B levels
Genetic approaches: siRNA knockdown, CRISPR/Cas9 gene editing, or overexpression systems to modulate TNFRSF10B levels
Combination studies: Testing TNFRSF10B agonists with chemotherapeutic agents to identify synergistic interactions
These approaches help elucidate the mechanisms of TNFRSF10B-mediated apoptosis and identify potential therapeutic strategies targeting this pathway .
TNFRSF10B Human, Sf9 protein can be employed in various drug discovery applications:
High-throughput screening: Development of binding assays to identify small molecules that modulate TNFRSF10B activity
Structure-based drug design: Using purified protein for crystallography studies to determine three-dimensional structure
Antibody development: Generation of therapeutic antibodies targeting TNFRSF10B
TRAIL mimetics testing: Evaluation of synthetic TRAIL variants or peptide mimetics for receptor activation
Combination therapy optimization: Identifying compounds that sensitize cancer cells to TRAIL-induced apoptosis
The recombinant protein serves as a valuable tool for these applications due to its high purity and consistent activity profile .
TNFRSF10B genetic variants can affect protein function through several mechanisms, although the precise functional impacts are still being elucidated. In silico analyses have revealed that the rs1047275 locus is located in binding sites for miR-379 and miR-473, potentially regulating TNFRSF10B expression. These microRNAs have been shown to play roles in drug response in lung cancer and colon cancer metastasis. If TNFRSF10B SNPs functionally impact protein expression or activity, this could result in deficient apoptotic responses in tumor cells, providing a tumor survival advantage and ultimately adversely impacting patient survival. Allelic losses in the chromosomal region (8p21–23) harboring TNFRSF10B have been reported as frequent events in several cancers including lung cancer, suggesting that attenuated TNFRSF10B/DR5-induced apoptosis, possibly due to genetic variations, is associated with cancer development and progression .
Researchers face several technical challenges when working with TNFRSF10B:
Protein stability: Death receptors like TNFRSF10B can be unstable and lose activity during storage or handling
Oligomerization requirements: Effective activation often requires receptor clustering, which can be difficult to replicate in vitro
Cell type specificity: Response to TNFRSF10B activation varies greatly between cell types
Background apoptosis: Distinguishing receptor-specific from non-specific cell death in experimental systems
Reproducibility issues: Variations in glycosylation patterns between protein batches can affect functional studies
Addressing these challenges requires careful experimental design, appropriate controls, and standardized protocols to ensure reproducible and meaningful results .
TNFRSF10B is emerging as a potential germline biomarker for personalized cancer treatment strategies. Studies have demonstrated that specific TNFRSF10B polymorphisms and haplotypes correlate with survival outcomes in NSCLC patients, particularly in relation to treatment response. The discovery that risk haplotypes exhibited increased mortality in surgical-only patients but not in patients receiving adjuvant chemotherapy suggests that TNFRSF10B genetic testing could help identify which early-stage patients would benefit from adjuvant chemotherapy. This represents a shift toward using germline biomarkers, rather than just tumor markers, for treatment stratification. Ongoing research is exploring the predictive value of TNFRSF10B variants across different cancer types, treatment regimens, and patient populations to develop more personalized therapeutic approaches .
Advanced techniques for studying TNFRSF10B signaling complex formation include:
Proximity ligation assays: Detecting protein-protein interactions in situ with high sensitivity
FRET/BRET techniques: Measuring real-time interactions between TNFRSF10B and adapter proteins
Mass spectrometry-based proteomics: Identifying components of the death-inducing signaling complex (DISC)
Super-resolution microscopy: Visualizing receptor clustering and complex formation at the nanoscale level
Hydrogen-deuterium exchange mass spectrometry: Mapping protein interaction interfaces
These methods help elucidate the dynamics and composition of the signaling complexes formed upon TNFRSF10B activation, which is crucial for understanding receptor functionality in normal and disease states .
TNFRSF10B contains an intracellular death domain, which is crucial for its role in apoptosis (programmed cell death). The receptor is activated by binding to the tumor necrosis factor-related apoptosis-inducing ligand (TNFSF10/TRAIL/APO-2L) . Upon activation, TNFRSF10B transduces an apoptosis signal, leading to the formation of the death-inducing signaling complex (DISC). This complex recruits and activates caspase-8, initiating a cascade of caspases that mediate apoptosis .
TNFRSF10B plays a significant role in regulating apoptosis, which is essential for maintaining cellular homeostasis and preventing tumor formation. Mutations or deregulation of this gene can lead to various cancers, including squamous cell carcinoma of the head and neck . The receptor’s ability to induce apoptosis makes it a potential target for cancer therapies, as activating TNFRSF10B can selectively induce death in cancer cells while sparing normal cells .
The recombinant form of TNFRSF10B, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain containing 394 amino acids and has a molecular mass of approximately 43.9 kDa . This recombinant protein is used in various research applications, including studying the mechanisms of apoptosis and developing potential therapeutic agents .
Recombinant TNFRSF10B is valuable in research for understanding the pathways involved in apoptosis and cancer progression. It is also used in cytotoxicity assays to measure its ability to inhibit the growth of cancer cells. The recombinant protein’s biological activity is measured by its ability to inhibit cytotoxicity in human T lymphocytes, with an effective dose (ED50) of less than or equal to 5 ng/ml when used with TRAIL .