TNFRSF10D is encoded by the TNFRSF10D gene located at chromosome 8p21.3. It is a type I transmembrane protein with three functional domains:
Extracellular TRAIL-binding domain: Binds TRAIL ligand.
Transmembrane domain: Anchors the receptor to the cell membrane.
Truncated cytoplasmic death domain: Lacks apoptotic signaling capacity .
Key aliases include CD264, DCR2, and TRUNDD.
TNFRSF10D is typically produced in insect cells (e.g., Sf9 Baculovirus) as a glycosylated polypeptide chain. Key features include:
Neutralizes TRAIL-mediated apoptosis in Jurkat T-cells (ED₅₀ ≤10 ng/ml) .
Acts as a decoy by binding TRAIL without triggering downstream apoptotic cascades .
TNFRSF10D interacts with key components of the TRAIL pathway:
Apoptosis Inhibition: Prevents TRAIL-induced cell death by blocking DR4/DR5 signaling .
NF-κB Activation: Conflicting reports exist on its role in NF-κB pathway modulation .
TNFRSF10D’s overexpression is linked to tumor resistance to TRAIL-based therapies. Key findings include:
Lung Cancer: No significant methylation association observed in NSCLC tissues vs. non-tumor controls .
Other Cancers:
| Cancer Type | Methylation Status | Association |
|---|---|---|
| Lung Cancer | No change in TNFRSF10D | No significant link to NSCLC |
| Breast Cancer | Hypermethylation | Silences expression, promotes survival |
TNFRSF10D is expressed in multiple human tissues and cell lines, including granulocytes and epithelial cells .
Key Expression Data:
| Dataset | Key Findings |
|---|---|
| BioGPS Tissue Atlas | High expression in blood granulocytes and lung tissues . |
| CCLE Cell Lines | Variable expression across cancer cell lines (e.g., leukemia, lung) . |
Antibodies: PE-conjugated monoclonal antibodies (e.g., Clone #104918) enable flow cytometry detection in granulocytes .
Neutralizing Assays: Recombinant TNFRSF10D protein is used to block TRAIL activity in vitro .
TRAIL Receptor-4, also known as TNFRSF10D in humans, belongs to the TNF-receptor superfamily. TNFRSF10D is characterized by a truncated cytoplasmic death domain, an extracellular TRAIL-binding domain, and a transmembrane domain. This protein has the ability to inhibit TRAIL-induced apoptosis in cells expressing TRAIL R1 and/or TRAIL R2.
Produced using Sf9 Baculovirus cells, TNFRSF10D is a single, glycosylated polypeptide chain consisting of 395 amino acids (specifically, residues 56-211a.a.). It has a molecular mass of 73.8kDa. However, on SDS-PAGE, the apparent molecular size may fall within the range of 40-57kDa.
TNFRSF10D is expressed with a C-terminal 239 amino acid hIgG-His tag and purified through proprietary chromatographic techniques.
The TNFRSF10D protein solution is supplied at a concentration of 0.5mg/ml and is prepared in a buffer consisting of phosphate buffered saline (pH 7.4) with 10% glycerol.
SDS-PAGE analysis indicates a purity exceeding 90.0%.
Biological activity is assessed through a neutralizing assay employing Jurkat human T lymphocytes. The ED50, representing the effective concentration at which 50% neutralization is achieved, is less than or equal to 10 ng/ml in the presence of 2 ng/ml TRAIL.
Tumor necrosis factor receptor superfamily member 10D, CD264, DCR2, TRAIL-R4, TRAILR4, TRUNDD, Decoy receptor 2, TNF-related apoptosis-inducing ligand receptor 4, TRAIL receptor 4, TRAIL receptor with a truncated death domain.
ATIPRQDEVP QQTVAPQQQR RSLKEEECPA GSHRSEYTGA CNPCTEGVDY TIASNNLPSC LLCTVCKSGQ TNKSSCTTTR DTVCQCEKGS FQDKNSPEMC RTCRTGCPRG MVKVSNCTPR SDIKCKNESA ASSTGKTPAA EETVTTILGM LASPYHVEPK SCDKTHTCPP CPAPELLGGP
SVFLFPPKPK DTLMISRTPE VTCVVVDVSH EDPEVKFNWY VDGVEVHNAK TKPREEQYNS TYRVVSVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK AKGQPREPQV YTLPPSRDEL TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ
QGNVFSCSVM HEALHNHYTQ KSLSLSPGKH HHHHH.
TNFRSF10D is known by several names in scientific literature, including TRAIL Receptor-4 (TRAIL-R4), Decoy receptor 2 (DCR2), TNF-related apoptosis-inducing ligand receptor 4, TRUNDD, and CD264. It belongs to the tumor necrosis factor receptor superfamily and is characterized as a TRAIL receptor with a truncated death domain . When conducting literature searches, researchers should use multiple search terms to ensure comprehensive coverage of relevant publications.
TNFRSF10D contains three key structural components: a truncated cytoplasmic death domain, an extracellular TRAIL-binding domain, and a transmembrane domain . The truncated nature of its death domain is particularly significant as it differentiates TNFRSF10D from other TRAIL receptors and explains its functional role. In recombinant form, human TNFRSF10D protein typically consists of 395 amino acids (spanning positions 56-211 of the native sequence) with a molecular mass of approximately 73.8 kDa, though it may appear at 40-57 kDa on SDS-PAGE due to glycosylation patterns .
TNFRSF10D functions primarily as a decoy receptor that can prevent TRAIL-mediated apoptosis on cells expressing TRAIL R1 and/or TRAIL R2 . Unlike the death-inducing TRAIL receptors, TNFRSF10D can bind TRAIL ligand but cannot transmit the apoptotic signal due to its truncated death domain. In experimental settings, Recombinant Human TRAIL R4/TNFRSF10D Fc Chimera has been shown to inhibit Recombinant Human TRAIL/TNFSF10-induced cytotoxicity in the L-929 mouse fibroblast cell line in a dose-dependent manner . This inhibitory effect can be neutralized by Human TRAIL R4/TNFRSF10D Monoclonal Antibody, with typical ND50 values ranging from 0.3-1.8 μg/mL .
Several validated methods exist for detecting TNFRSF10D expression:
Flow Cytometry: Flow cytometry represents an effective approach for detecting TNFRSF10D/DcR2 expression on cell surfaces. Research has employed this technique to measure expression levels on various cell lines including Colo205 and HCT116 cells . Detection typically employs anti-FLAG-PE for visualization of bound molecules. Flow cytometry allows researchers to simultaneously assess TNFRSF10D alongside other markers such as EGFR, HER2, HER3, and EpCAM.
Sandwich Immunoassay: This method provides quantitative detection of TNFRSF10D. Optimal antibody dilutions should be determined by each laboratory for specific applications .
Neutralizing Assays: Biological activity of TNFRSF10D can be measured in a neutralizing assay using Jurkat human T lymphocytes. The ED50 for TNFRSF10D effect is typically less than or equal to 10 ng/mL in the presence of 2 ng/mL TRAIL .
For TNFRSF10D DNA methylation analysis in clinical samples, researchers have established the following protocol:
DNA Extraction from FFPE Tissues: Extract DNA from small punch-biopsies from formalin-fixed paraffin-embedded (FFPE) tissue samples.
Bisulfite Conversion: Perform bisulfite conversion on extracted DNA to preserve methylation information.
MethyLight PCR: Analyze the methylation status using MethyLight PCR technique .
This methodology has been successfully applied to melanoma specimens, demonstrating that TNFRSF10D DNA-methylation analysis from small tissue-punches from archival FFPE is feasible and provides valuable prognostic information . The approach is particularly valuable as it can be conducted on readily available archived specimens, enhancing its utility in retrospective studies where fresh tissue is unavailable.
When designing experiments to study TNFRSF10D's inhibitory effects on TRAIL-induced apoptosis, researchers should consider:
Cell Line Selection: Choose cell lines with known TRAIL sensitivity, such as L-929 mouse fibroblasts or Jurkat human T lymphocytes .
Use of Metabolic Inhibitors: Consider including metabolic inhibitors such as actinomycin D, which can enhance TRAIL sensitivity in certain cell types .
Dose-Response Analysis: Implement a dose-dependent experimental design where Recombinant Human TRAIL R4/TNFRSF10D Fc Chimera concentrations are varied against a fixed concentration of Recombinant Human TRAIL/TNFSF10. For example, studies have used 90 ng/mL of TRAIL R4/TNFRSF10D Fc Chimera against 20 ng/mL of TRAIL/TNFSF10 .
Neutralization Assessment: Include conditions where Human TRAIL R4/TNFRSF10D Monoclonal Antibody is used to neutralize the inhibitory effect of TNFRSF10D, with typical ND50 values of 0.3-1.8 μg/mL .
Detection Methods: Employ multiple readouts for apoptosis, including cell viability assays, caspase activation measurements, and flow cytometry for apoptotic markers.
TNFRSF10D DNA methylation has emerged as a significant prognostic marker in cancer, particularly in melanoma. Research indicates:
This data suggests that TNFRSF10D methylation status might serve as a more robust prognostic indicator than traditional clinicopathological parameters in melanoma patients.
The comparison between TNFRSF10D methylation and other established prognostic factors reveals important insights:
While the search results don't directly address personalized treatment approaches based on TNFRSF10D methylation, several implications can be inferred:
Risk Stratification: Patients could be stratified into risk groups based on TNFRSF10D methylation status, potentially allowing for more intensive surveillance or adjuvant therapy for high-risk patients (those with methylated TNFRSF10D).
Treatment Selection: The strong prognostic value of TNFRSF10D methylation suggests it might help identify patients more likely to benefit from aggressive therapies or novel treatment approaches.
Combination with Interferon Therapy: The data indicates a potential interaction between interferon alpha therapy and prognostic outcomes. This suggests that TNFRSF10D methylation status might help identify patients who would benefit most from interferon therapy .
Epigenetic Therapy Potential: Given that DNA methylation is potentially reversible, patients with methylated TNFRSF10D might benefit from epigenetic therapies that could restore normal TNFRSF10D expression.
The exact molecular mechanisms connecting TNFRSF10D methylation to cancer progression are not explicitly detailed in the search results, but based on its function, several mechanisms can be proposed:
Apoptosis Resistance: TNFRSF10D functions as a decoy receptor that can inhibit TRAIL-mediated apoptosis . Methylation of TNFRSF10D could potentially silence its expression, altering the balance between death receptors and decoy receptors, thereby affecting cellular sensitivity to TRAIL-mediated apoptosis.
Immune Evasion: TRAIL is produced by immune cells and plays a role in tumor surveillance. Alterations in TNFRSF10D expression via methylation could impact how cancer cells respond to immune-mediated TRAIL signaling.
Cell Survival Signaling: Beyond its role in apoptosis, TRAIL receptor signaling has been implicated in non-apoptotic pathways including NF-κB activation and proliferation. TNFRSF10D methylation might influence these alternative signaling outputs.
Interaction with Other Prognostic Factors: The correlation between TNFRSF10D methylation and other prognostic factors like age, Clark level, and mitotic rate suggests potential interactions with fundamental processes governing melanoma biology .
Future research should focus on elucidating these mechanisms to better understand the biological basis of TNFRSF10D methylation's prognostic impact.
Heterogeneity in TNFRSF10D expression can significantly impact both experimental results and clinical interpretations:
Cellular Heterogeneity: Expression levels of TNFRSF10D can vary between different cell types and even within a single tumor. Flow cytometry studies have shown variable expression levels of TRAIL receptors including TNFRSF10D across different cell lines and treatments . This heterogeneity might affect the interpretation of bulk tissue analysis.
Treatment-Induced Changes: Research has demonstrated that treatment with agents like BZB (250 ng/ml for 16h) can alter TNFRSF10D expression levels . This dynamic regulation means that expression at a single timepoint may not represent the full biological context.
Technical Variability: Different detection methods (flow cytometry, immunoassays, etc.) might yield different results regarding TNFRSF10D expression levels, necessitating method standardization for cross-study comparisons.
Methylation Heterogeneity: The binary classification of TNFRSF10D as "methylated" versus "unmethylated" may oversimplify a more complex pattern of partial or heterogeneous methylation within a tumor sample.
Researchers should account for these sources of heterogeneity through approaches such as single-cell analysis, spatial profiling, longitudinal sampling, and robust technical replicates.
When faced with contradictory results in TNFRSF10D research, researchers should consider:
Researchers should explicitly address these considerations when designing studies, analyzing data, and interpreting conflicting literature in the field.
Several innovative approaches could enhance TNFRSF10D's clinical utility:
Liquid Biopsy Development: Developing methods to detect TNFRSF10D methylation in circulating tumor DNA from blood samples could allow for non-invasive longitudinal monitoring of patients.
Integrated Multi-Marker Panels: Combining TNFRSF10D methylation with other molecular markers, immunohistochemical features, and clinical parameters into integrated prognostic models might improve predictive accuracy.
Artificial Intelligence Applications: Machine learning approaches could help identify subtle patterns in TNFRSF10D methylation levels or distribution that correlate with outcomes and might not be apparent through conventional statistical analysis.
Standardized Assays: Development of clinically validated, standardized assays for TNFRSF10D methylation would facilitate implementation in diagnostic laboratories and enable cross-study comparisons.
Spatial Methylation Analysis: Techniques that preserve spatial information about TNFRSF10D methylation within the tumor microenvironment could provide insights into the relationship between methylation patterns and local tumor-immune interactions.
To better understand the functional impact of TNFRSF10D methylation, researchers should consider:
CRISPR-Based Epigenetic Editing: Using CRISPR-Cas9 coupled with DNA methyltransferases or demethylases to specifically modify TNFRSF10D methylation status in cell lines would allow for controlled experiments examining the direct consequences of methylation changes.
Patient-Derived Xenografts: Creating xenograft models from patient tumors with known TNFRSF10D methylation status would enable in vivo studies of tumor behavior and treatment response.
Single-Cell Analysis: Applying single-cell techniques to analyze TNFRSF10D methylation, expression, and functional outcomes would help address the impact of intratumoral heterogeneity.
Dynamic Monitoring: Longitudinal studies tracking changes in TNFRSF10D methylation during disease progression and in response to therapy would clarify its role in disease evolution.
Mechanistic Studies: Detailed biochemical and cell biological investigations of how TNFRSF10D methylation affects protein expression, localization, and interaction with TRAIL and other signaling partners would illuminate the molecular basis of its clinical significance.
TNFRSF10D research could contribute to therapeutic innovation through several avenues:
Epigenetic Drug Development: Understanding the mechanisms and consequences of TNFRSF10D methylation could inform the development of targeted epigenetic therapies aimed at restoring normal TNFRSF10D expression patterns.
Combination Therapy Optimization: The prognostic value of TNFRSF10D methylation in the context of treatments like interferon alpha therapy suggests potential for optimizing combination regimens based on methylation status .
TRAIL Pathway Modulation: Given TNFRSF10D's role as a decoy receptor in TRAIL signaling, research could inform strategies to modulate the TRAIL pathway in cancer, potentially enhancing the efficacy of TRAIL-based therapeutics by accounting for TNFRSF10D status.
Immunotherapy Biomarkers: Exploring the relationship between TNFRSF10D methylation and response to immunotherapies could yield predictive biomarkers, particularly given TRAIL's role in immune surveillance.
Precision Medicine Approaches: The strong prognostic value of TNFRSF10D methylation suggests its potential utility in precision medicine algorithms for treatment selection and intensity, particularly in melanoma patients.
TRAILR4 is a type 1 transmembrane protein that lacks a functional death domain, which is why it is referred to as a “decoy” receptor . Unlike other TRAIL receptors, such as death receptor 4 (DR4) and death receptor 5 (DR5), TRAILR4 does not induce apoptosis. Instead, it acts as a regulatory receptor that can inhibit TRAIL-induced apoptosis by competing with DR4 and DR5 for TRAIL binding .
TRAILR4 has garnered significant interest in cancer research due to its ability to modulate the apoptotic response in tumor cells. TRAIL, the ligand for TRAILR4, selectively induces apoptosis in cancer cells while sparing normal cells . This selective induction of cell death makes TRAIL and its receptors promising targets for cancer therapy .
Recombinant human TRAILR4 is produced using recombinant DNA technology, which involves inserting the gene encoding TRAILR4 into a suitable expression system, such as a bacterial or mammalian cell line. The recombinant protein is then purified and used for various research and therapeutic applications .
Recombinant TRAILR4 is used in research to study its role in apoptosis and cancer biology. It is also utilized in drug development to screen for potential therapeutic agents that can modulate TRAILR4 activity . Additionally, recombinant TRAILR4 can be used in diagnostic assays to measure the levels of TRAILR4 in biological samples .