TNFRSF10C plays a critical role in modulating programmed cell death by functioning as a decoy receptor in the TRAIL-mediated apoptotic pathway . By binding TRAIL without initiating apoptotic signaling, TNFRSF10C effectively competes with death-inducing receptors TRAIL-R1 (TNFRSF10A) and TRAIL-R2 (TNFRSF10B) . This competitive binding prevents TRAIL from activating its pro-apoptotic receptors, thereby protecting cells from TRAIL-induced cell death .
The expression of TNFRSF10C has been shown to protect cells bearing TRAIL-R1 and/or TRAIL-R2 from TRAIL-induced apoptosis, highlighting its role as an antagonistic receptor . This protective mechanism is particularly significant in normal tissues, where TNFRSF10C expression may confer selective resistance to TRAIL-mediated apoptosis .
TNFRSF10C exhibits a distinctive expression pattern with significant biological implications. It is expressed in many normal tissues but is notably absent in most cancer cell lines . This differential expression pattern may partially explain the selective sensitivity of cancer cells to TRAIL-induced apoptosis .
Research has identified TNFRSF10C as a p53-regulated DNA damage-inducible gene, suggesting its expression is modulated in response to cellular stress and DNA damage . This regulatory mechanism potentially links TNFRSF10C expression to cellular surveillance systems and defense mechanisms against malignant transformation.
Hemizygous deletion of the TNFRSF10C gene is frequently observed in various cancer types. Studies have documented TNFRSF10C deletion in 74.5% of prostate tumors, representing a significant genetic alteration in this malignancy . This high frequency of deletion correlates with the gene's location in a chromosomal region commonly lost during cancer development.
In addition to genetic deletion, epigenetic silencing through promoter CpG island hypermethylation represents another major mechanism of TNFRSF10C inactivation in cancer . Research has documented TNFRSF10C promoter hypermethylation in numerous malignancies with varying frequencies:
Cancer Type | Frequency of TNFRSF10C Promoter Hypermethylation |
---|---|
Prostate cancer | 78.0% |
Neuroblastoma | 21% |
Primary breast cancer | 33% |
Primary lung cancer | 16% |
Malignant mesotheliomas | 63% |
Ependymoma | 50% |
Choroid plexus papillomas | 50% |
Table 1: Frequency of TNFRSF10C promoter hypermethylation across various cancer types
A comprehensive study of prostate cancer revealed that in 94.9% of tumors (56 out of 59), TNFRSF10C was either hemizygously deleted or its promoter hypermethylated . This extraordinarily high frequency of inactivation suggests that TNFRSF10C silencing may play a critical role in prostate cancer development and progression.
Furthermore, this inactivation was directly correlated with decreased mRNA expression of the gene in clinical specimens, confirming the functional consequences of these genetic and epigenetic alterations . Experimental demethylation of the TNFRSF10C promoter in the prostate cancer cell line PC3 resulted in transcriptional re-activation of the gene, establishing a causal relationship between promoter methylation and gene silencing .
Recombinant human TNFRSF10C is typically produced using mammalian expression systems to ensure proper folding and post-translational modifications, particularly glycosylation, which is critical for the protein's function . Human cell lines such as HEK293 cells are commonly employed as expression hosts to maintain native protein characteristics .
The production of recombinant TNFRSF10C generally involves expressing the extracellular domain of the protein, which contains the TRAIL-binding region. Commercial recombinant TNFRSF10C proteins often express a specific segment of the protein, such as the region from Alanine-26 to Alanine-221, representing the functional extracellular portion of the receptor .
Recombinant TNFRSF10C proteins typically incorporate affinity tags to facilitate purification and detection . Common tagging strategies include:
Tag Type | Position | Function |
---|---|---|
Histidine (His) tag | C-terminal or N-terminal | Facilitates purification via metal affinity chromatography |
Fc fusion tag | C-terminal | Enhances stability and facilitates purification with protein A/G |
Myc tag | C-terminal | Enables immunodetection with anti-Myc antibodies |
Table 2: Common tagging strategies for recombinant TNFRSF10C proteins
These tags enable efficient purification through affinity chromatography, resulting in high-purity preparations. Commercial recombinant TNFRSF10C proteins typically achieve purity levels exceeding 95% as determined by SDS-PAGE analysis .
The molecular weight of recombinant TNFRSF10C typically ranges from 38-58 kDa, though this can vary depending on the expression system, the exact boundaries of the expressed region, and the nature of any fusion tags . The observed molecular weight may also be influenced by glycosylation patterns, which can differ based on the expression host.
The primary functional property of recombinant TNFRSF10C is its ability to bind TRAIL with high affinity . This binding capacity is essential for its biological role as a decoy receptor and is the key property evaluated when assessing the activity of recombinant preparations. Functional assays typically measure the protein's binding ability in a quantitative ELISA format, with active preparations demonstrating dose-dependent TRAIL binding .
Recombinant TNFRSF10C serves as a valuable tool for investigating TRAIL signaling pathways and their regulation. It enables researchers to study:
The competitive binding dynamics between different TRAIL receptors
The protective mechanisms against TRAIL-induced apoptosis
The structural basis of TRAIL-receptor interactions
The differential sensitivity of cancer versus normal cells to TRAIL
These studies contribute to our understanding of how TRAIL signaling is regulated in normal tissues and how dysregulation of this pathway may contribute to cancer development.
The frequent inactivation of TNFRSF10C in various cancers has stimulated research into its potential role as a tumor suppressor and its utility in cancer diagnostics and therapeutics:
Biomarker Development: TNFRSF10C promoter methylation status may serve as a potential biomarker for cancer detection or prognosis
Epigenetic Therapy: Targeting the reversal of TNFRSF10C promoter methylation to restore its expression in cancer cells
Selective Cancer Targeting: Developing strategies that exploit the differential expression of TNFRSF10C between normal and cancer cells
Drug Screening: Using recombinant TNFRSF10C in high-throughput screens to identify compounds that modulate TRAIL receptor interactions
Recombinant TNFRSF10C enables detailed investigation of protein-protein interactions within the TRAIL signaling network. Research has identified several key protein partners of TNFRSF10C:
Protein Partner | Interaction Type | Functional Significance |
---|---|---|
TNFSF10 (TRAIL) | Ligand-receptor binding | Primary interaction; competitive binding with death receptors |
TNFRSF10A (TRAIL-R1) | Indirect competition | Compete for same ligand (TRAIL) |
TNFRSF10B (TRAIL-R2) | Indirect competition | Compete for same ligand (TRAIL) |
TNFRSF10D (TRAIL-R4) | Functional redundancy | Both serve as decoy receptors for TRAIL |
Table 3: Key protein interaction partners of TNFRSF10C
The understanding of TNFRSF10C's role in cancer has prompted exploration of potential therapeutic approaches:
Development of selective TRAIL variants that can bypass decoy receptors while maintaining activity toward death receptors
Epigenetic drugs targeting the reversal of TNFRSF10C silencing in cancer cells
Diagnostic applications using TNFRSF10C methylation or deletion status as biomarkers
Integration of TNFRSF10C into systems biology frameworks will help elucidate:
The complex interplay between pro- and anti-apoptotic TRAIL receptors
Tissue-specific regulation of TRAIL sensitivity
Evolutionary conservation and divergence of TRAIL signaling components
TNFRSF10C, also known as TRAIL-R3 or DcR1 (Decoy Receptor 1), functions as a decoy receptor for the TNF-related apoptosis-inducing ligand (TRAIL). Unlike functional death receptors, TNFRSF10C binds to TRAIL but lacks the death domain necessary for apoptosis signaling. Consequently, it inhibits TRAIL-induced apoptosis by competing with death receptors (DR4 and DR5) for TRAIL binding . This competitive inhibition creates a regulatory mechanism for TRAIL-mediated cell death pathways, allowing cells to modulate their sensitivity to apoptotic signals.
When designing experiments to study TNFRSF10C function, researchers should consider:
Co-expression levels of other TRAIL receptors (DR4, DR5, and DcR2)
The ratio of decoy to functional receptors in the specific cell type
Post-translational modifications that might affect binding affinity
Tissue-specific expression patterns that may influence experimental outcomes
TNFRSF10C possesses unique structural characteristics that distinguish it from other TRAIL receptors:
It contains the extracellular TRAIL-binding domain
It lacks a functional intracellular death domain
It is anchored to the cell membrane through a glycosylphosphatidylinositol (GPI) linkage rather than a transmembrane domain
This structural arrangement allows TNFRSF10C to bind TRAIL without initiating apoptotic signaling cascades, effectively sequestering the ligand and preventing its interaction with death-domain-containing receptors.
When producing recombinant TNFRSF10C for research applications, several expression systems have been successfully employed, each with distinct advantages:
Expression System | Advantages | Limitations | Yield | Post-translational Modifications |
---|---|---|---|---|
E. coli | High yield, cost-effective, rapid production | Lacks glycosylation, potential improper folding | High | Minimal |
Mammalian cells (HEK293, CHO) | Native-like glycosylation, proper folding | Higher cost, slower production | Moderate | Extensive and native-like |
Insect cells (Sf9, High Five) | Proper folding, some glycosylation | Glycosylation pattern differs from human | High | Intermediate |
Yeast (P. pastoris) | High yield, some glycosylation | Hyperglycosylation may occur | High | Different pattern than human |
For functional studies requiring proper receptor-ligand interactions, mammalian expression systems are generally recommended due to their ability to produce correctly folded and glycosylated proteins. For structural studies where glycosylation is less critical, bacterial systems may be suitable after optimization of solubilization and refolding protocols.
Purification of active recombinant TNFRSF10C requires careful consideration of protein stability and functional integrity. A multi-step purification approach is typically recommended:
Initial capture: Affinity chromatography using His-tag, FLAG-tag, or immunoaffinity approaches
Intermediate purification: Ion exchange chromatography to separate charged variants
Polishing: Size exclusion chromatography to remove aggregates and ensure monodispersity
Critical considerations include:
Maintaining physiological pH (7.2-7.4) throughout purification to preserve native conformation
Including stabilizing agents (glycerol 10-15%, low concentrations of non-ionic detergents)
Minimizing freeze-thaw cycles which can lead to activity loss
Confirming biological activity through TRAIL binding assays post-purification
TNFRSF10C promoter hypermethylation has been observed in multiple cancer types and correlates with disease progression. In non-small cell lung cancer (NSCLC), methylation levels of TNFRSF10C in tumor tissues are significantly higher compared to distant non-tumor tissues (P=0.013) . Similar hypermethylation patterns have been documented in glioblastoma, prostate cancer, and breast cancer .
Methylation analysis reveals interesting sex-specific and smoking status-dependent patterns:
Male patients show higher TNFRSF10C methylation in tumor tissues compared to females (P=0.013)
Non-smokers exhibit significant differences in TNFRSF10C methylation between tumor and non-tumor tissues (P=0.031)
In male non-smokers, a unique pattern of hypomethylation was observed (P=0.03)
These findings suggest that TNFRSF10C methylation status could serve as a potential biomarker for cancer diagnosis and might explain differential responses to TRAIL-based therapies across patient demographics.
Several methodologies have been employed to analyze TNFRSF10C methylation with varying degrees of sensitivity and specificity:
Method | Sensitivity | Quantitative | Resolution | Sample Requirements | Advantages |
---|---|---|---|---|---|
Quantitative Methylation-Specific PCR (qMSP) | High | Yes | Region-specific | Low DNA input (50-100ng) | High throughput, cost-effective |
Bisulfite Sequencing | High | Yes | Single CpG site | Moderate DNA input (200-500ng) | Comprehensive CpG analysis |
Methylation Arrays | Moderate | Yes | Predefined CpG sites | Moderate DNA input (250-500ng) | Genome-wide perspective |
Pyrosequencing | High | Yes | Single CpG site | Low DNA input (50-100ng) | Quantitative, accurate |
For TNFRSF10C specifically, qMSP has been successfully employed using primers targeting CG-rich regions in the promoter. The primer sequences reported in the literature are:
When designing methylation studies for TNFRSF10C, researchers should consider:
Including both tumor and matched non-tumor tissues when possible
Stratifying analysis by relevant clinical parameters (sex, smoking status, etc.)
Validating methylation findings with gene expression analysis
Confirming functional relevance through in vitro demethylation experiments
The relationship between TNFRSF10C promoter methylation and gene expression has been demonstrated through multiple lines of evidence:
Inverse correlation between methylation and expression: Analysis of lung cancer data from cBioPortal database revealed that TNFRSF10C methylation levels were negatively correlated with its mRNA expression (r=-0.379, P=0.008) .
Demethylation experiments: Treatment of lung cancer cell lines (A549 and NCI-H1299) with 5′-aza-deoxycytidine resulted in significant increases in TNFRSF10C mRNA expression:
Promoter activity assays: Dual luciferase reporter assays using the TNFRSF10C promoter fragment (-121 to +363 bp) demonstrated enhanced transcriptional activity (fold-change=1.570, P=0.032) , confirming this region's role in regulating gene expression.
Mechanistically, methylation of CpG islands in the TNFRSF10C promoter likely interferes with the binding of transcription factors necessary for gene expression. This epigenetic silencing mechanism appears to be particularly relevant in specific cancer contexts and patient subgroups.
The dysregulation of TNFRSF10C through epigenetic mechanisms has significant implications for TRAIL-based cancer therapeutics:
Hypermethylation and silencing of TNFRSF10C could potentially enhance cancer cell sensitivity to TRAIL-induced apoptosis by reducing the decoy receptor's competitive inhibition .
Conversely, in some contexts, TNFRSF10C silencing might represent a mechanism through which tumors disrupt normal apoptotic signaling, contributing to immune evasion.
Patient stratification considerations: The observed sex-specific and smoking status-dependent methylation patterns suggest that response to TRAIL-based therapies might vary across patient demographics.
When designing TRAIL-based therapeutic studies, researchers should consider:
Evaluating TNFRSF10C methylation status as a potential predictive biomarker
Assessing the ratio of functional to decoy TRAIL receptors in target tissues
Exploring combination approaches using demethylating agents with TRAIL-based therapies
Investigating tissue-specific and demographic-specific responses
Selecting appropriate cell models is critical for studying TNFRSF10C function in cancer contexts:
Cell Type | TNFRSF10C Characteristics | Applications | Limitations |
---|---|---|---|
A549 (lung adenocarcinoma) | Methylated TNFRSF10C, responsive to demethylation | Methylation studies, expression regulation | May not represent all NSCLC subtypes |
NCI-H1299 (lung carcinoma) | Methylated TNFRSF10C, responsive to demethylation | Methylation-expression correlation studies | p53-null, which may affect apoptotic pathways |
293T (embryonic kidney) | High transfection efficiency | Promoter activity assays, protein production | Not a cancer cell line |
Primary patient-derived cells | Clinically relevant methylation patterns | Translational research, precision medicine | Limited availability, heterogeneity |
When designing experiments:
Include multiple cell lines to account for heterogeneity
Consider the baseline expression of all TRAIL receptors
Verify methylation status of the specific cell line batch
Assess the impact of culture conditions on TNFRSF10C expression and methylation
Traditional bulk analysis methods may obscure important cellular heterogeneity in TNFRSF10C expression and methylation. Single-cell approaches offer several advantages:
Resolving intratumoral heterogeneity:
Identifying subpopulations with differential TNFRSF10C expression
Correlating single-cell methylation with expression at the individual cell level
Mapping TRAIL sensitivity to receptor expression patterns
Methodological considerations:
Single-cell RNA sequencing can reveal expression patterns across tumor populations
Single-cell ATAC-seq can identify chromatin accessibility at the TNFRSF10C locus
Single-cell bisulfite sequencing, while technically challenging, can directly assess methylation
Data integration strategies:
Combining single-cell RNA-seq with protein-level measurements (CyTOF, CITE-seq)
Integrating spatial information to understand the tumor microenvironment context
Correlating with clinical outcomes to identify prognostically relevant subpopulations
The potential of TNFRSF10C methylation as a clinical biomarker is supported by several lines of evidence:
Cancer-specific hypermethylation: TNFRSF10C shows differential methylation between tumor and non-tumor tissues across multiple cancer types .
Non-invasive detection potential: While tissue-based methylation analysis provides the current standard, investigating TNFRSF10C methylation in liquid biopsies (circulating tumor DNA, exosomes) could provide less invasive diagnostic opportunities .
Demographic considerations: The observed sex-specific and smoking status-dependent methylation patterns suggest potential for developing targeted screening approaches.
For clinical biomarker development, researchers should consider:
Standardizing methylation detection methodologies for clinical application
Establishing robust cutoff values for positive/negative results
Conducting large-scale validation studies across diverse patient populations
Evaluating the combination of TNFRSF10C methylation with other biomarkers for improved sensitivity and specificity
Several therapeutic strategies targeting the TRAIL/TNFRSF10C axis show promise for cancer treatment:
Epigenetic modifiers:
DNA methyltransferase inhibitors (5-azacytidine, decitabine) to reverse TNFRSF10C hypermethylation
Histone deacetylase inhibitors to enhance expression of silenced genes
TRAIL-based therapeutics:
Recombinant TRAIL or TRAIL-mimetic agents
Agonistic antibodies against death receptors (DR4, DR5)
Bispecific antibodies targeting both tumor antigens and TRAIL receptors
Combination approaches:
Demethylating agents + TRAIL to enhance sensitivity
Conventional chemotherapy + TRAIL for synergistic effects
Immune checkpoint inhibitors + TRAIL pathway activation
When designing therapeutic studies:
Characterize baseline TNFRSF10C methylation and expression
Consider sex and smoking status as potential modifiers of treatment response
Monitor changes in methylation status during treatment
Develop companion diagnostics for patient stratification
Recombinant TNFRSF10C presents several technical challenges that researchers should consider:
Stability issues:
Protein aggregation during storage
Loss of activity upon freeze-thaw cycles
Structural changes affecting TRAIL binding
Recommended solutions:
Addition of stabilizers: 10-15% glycerol, low concentrations of non-ionic detergents
Single-use aliquots to avoid freeze-thaw cycles
Storage at -80°C for long-term or 4°C for short-term (1-2 weeks)
Quality control testing of each batch for TRAIL binding activity
Functional validation approaches:
Competitive binding assays with labeled TRAIL
Surface plasmon resonance to measure binding kinetics
Cell-based assays measuring inhibition of TRAIL-induced apoptosis
Robust methylation analysis requires careful consideration of controls:
Technical controls:
Fully methylated and unmethylated DNA standards for qMSP calibration
Multiple primer sets targeting different CpG regions when possible
Non-bisulfite converted controls to verify conversion efficiency
Biological controls:
Matched tumor/normal tissue pairs from the same patient
Cell lines with known methylation status
Tissues from different demographic groups (considering sex and smoking status variables)
Validation approaches:
Correlation with gene expression data
Functional studies using demethylating agents
Promoter activity assays to confirm the relevance of specific CpG regions
When reporting methylation data, researchers should clearly describe:
The specific genomic region analyzed (including genome coordinates)
The methodology employed with detailed protocols
The quantification and normalization approaches
The statistical methods used for data analysis