Our Recombinant Human TNFSF10 (Tumor necrosis factor ligand superfamily member 10) is a high-quality protein designed for cancer research. TNFSF10, also known as Apo-2 ligand, Apo-2L, or TRAIL, is a key player in apoptosis induction and has shown promise as a potential therapeutic target in cancer treatment.
This tag-free recombinant protein is produced using an E. coli expression system, ensuring its purity of over 95% as determined by SDS-PAGE analysis. It covers a partial protein length spanning amino acids 115 to 281, providing a reliable tool for your cancer-related studies.
The activity of our TNFSF10 has been carefully evaluated. In a cytotoxicity assay using L-929 mouse fibroblast cells and the metabolic inhibitor actinomycin D, the effective dose (ED50) was found to be less than 50 ng/ml. Additionally, the product has been rigorously tested to have an endotoxin level of less than 1.0 EU/µg, ensuring its suitability for sensitive applications. It is supplied as a convenient lyophilized powder, ensuring stability and ease of use in your research experiments and applications.
Our Recombinant Human TNFSF10 (Tumor necrosis factor ligand superfamily member 10) is a premium quality protein meticulously designed for cancer research. TNFSF10, also known as Apo-2 ligand, Apo-2L, or TRAIL, plays a pivotal role in apoptosis induction and exhibits significant potential as a therapeutic target in cancer treatment.
This tag-free recombinant protein is produced using a robust E. coli expression system, ensuring a high purity exceeding 95%, as determined by SDS-PAGE analysis. It encompasses a partial protein sequence spanning amino acids 115 to 281, providing a reliable tool for your cancer-related investigations.
The activity of our TNFSF10 has been rigorously evaluated. In a cytotoxicity assay using L-929 mouse fibroblast cells and the metabolic inhibitor actinomycin D, the effective dose (ED50) was determined to be less than 50 ng/ml. Furthermore, the product has undergone comprehensive testing, resulting in an endotoxin level below 1.0 EU/µg, ensuring its suitability for sensitive applications. It is conveniently supplied as a lyophilized powder, guaranteeing stability and ease of use in your research experiments and applications.
TNFSF10, also known as TNF-related apoptosis-inducing ligand (TRAIL), Apo-2 ligand, or CD253, is a cytokine belonging to the tumor necrosis factor (TNF) ligand family. It functions as a homotrimeric type II transmembrane ligand that preferentially induces apoptosis in transformed and tumor cells while sparing normal cells .
Beyond its well-established role in apoptosis, recent evidence indicates TNFSF10 plays significant roles in:
Regulating antitumor immunity in cancer cells and the tumor microenvironment (TME)
Promoting granzyme B expression in cytotoxic T-cells
Potentially stimulating proliferation of regulatory T cells and M2 macrophages
TNFSF10 interacts with several members of the TNF receptor superfamily, including TNFRSF10A/TRAILR1, TNFRSF10B/TRAILR2, TNFRSF10C/TRAILR3, TNFRSF10D/TRAILR4, and possibly TNFRSF11B/OPG . Its apoptotic activity may be modulated by decoy receptors that cannot induce apoptosis.
For optimal storage and reconstitution of recombinant TNFSF10:
Storage conditions:
Store lyophilized protein at -20°C to -80°C
After reconstitution, prepare working aliquots to avoid repeated freeze-thaw cycles
For short-term use (up to one week), reconstituted protein can be stored at 4°C
For long-term storage, keep reconstituted protein at -20°C or -80°C
Reconstitution protocol:
Briefly centrifuge the vial to collect all material at the bottom
Reconstitute in sterile water or appropriate buffer (commonly PBS or 25mM Tris, 150mM NaCl, pH 7.5)
Gently mix by inverting the vial several times, avoid vigorous vortexing
Allow to stand for 5-10 minutes at room temperature before aliquoting
Filter through a 0.22μm filter if sterility is required for cell culture applications
Note that active recombinant TNFSF10 typically has an ED50 < 40 ng/mL, as measured by cell growth inhibitory assay using RPMI-8226 cells, corresponding to a specific activity of > 2.5 × 10^4 units/mg .
The differences between full-length and partial (commonly AA 114-281) recombinant TNFSF10 have important implications for research applications:
Full-length TNFSF10 (1-281):
Contains the cytoplasmic domain (AA 1-18), transmembrane domain (AA 18-28), and extracellular domain (AA 28-281)
More closely resembles native protein conformation
Often used for studying membrane-bound TRAIL functions
May provide context for studying interactions with the cellular membrane
Partial/soluble TNFSF10 (typically AA 114-281):
Contains only the receptor-binding domain
Higher specific activity in apoptosis assays
Better protein yield and stability in recombinant expression systems
Lower tendency to aggregate compared to full-length protein
Research has shown that the partial recombinant form (AA 114-281) retains full biological activity for inducing apoptosis through death receptors while offering superior stability characteristics . This explains why many commercial sources provide the partial active form rather than the full-length protein.
When designing TNFSF10-induced apoptosis assays, consider the following methodology:
Experimental design:
Cell selection: Use established TRAIL-sensitive (e.g., RPMI-8226, Jurkat) and TRAIL-resistant cell lines (e.g., some primary cells) as controls
Dose response: Test concentrations ranging from 1-1000 ng/mL of recombinant TNFSF10
Time course: Evaluate apoptosis at multiple timepoints (typically 4, 8, 12, 24 hours)
Combination studies: Consider testing TNFSF10 with sensitizing agents like proteasome inhibitors or chemotherapeutic drugs
Essential controls:
Untreated cells (negative control)
Known apoptosis inducer (e.g., staurosporine) as positive control
Heat-inactivated TNFSF10 (specificity control)
Pan-caspase inhibitor (e.g., Z-VAD-FMK) to confirm caspase-dependent mechanism
Measurement methods (use at least two):
Annexin V/PI staining with flow cytometry
Caspase 3/7 activity assays
DNA fragmentation analysis
PARP cleavage detection by western blot
Data should be presented showing both dose-response and time-course relationships, with statistical analysis of at least three independent experiments.
For comprehensive analysis of TNFSF10 expression:
mRNA expression analysis:
qRT-PCR: Use TaqMan or SYBR Green assays with properly validated primers
RNA-Seq: For genome-wide expression context
Analyze TNFSF10 in context of pathway activation
Correlate with expression of other immune genes
Protein expression analysis:
Western blot: For semi-quantitative analysis
Flow cytometry: For cell surface TNFSF10
Distinguishes between membrane-bound and intracellular forms
Use non-permeabilizing conditions for surface detection
ELISA: For soluble TNFSF10 in supernatants/serum
Commercial kits available with detection limits ~2-5 pg/mL
When analyzing TNFSF10 expression, it's critical to account for both membrane-bound and soluble forms, as metalloprotease-mediated shedding can affect detection . Additionally, distinguishing between basal expression and induction (e.g., by IFNs) provides important context for understanding regulatory mechanisms.
TNFSF10's role in antiviral immunity is distinct from yet complementary to its apoptotic function:
TNFSF10 in antiviral responses:
Expression is strongly induced by type I interferons (IFN-α, IFN-β) but minimally by TNF-α, despite being a TNF family member
Forms part of the innate immune response to viral infection
Correlates highly with expression of classical antiviral genes (DDX58/RIG-I, IFIH1/MDA-5, OAS1)
Contributes to poly(I:C)-induced apoptosis (mimicking viral RNA sensing)
Experimental evidence demonstrates that:
IFN-β dramatically increases TNFSF10 expression in triple-negative breast cancer (TNBC) cells
TNFSF10 expression correlates significantly with antiviral gene expression
TNFSF10 knockout reduces poly(I:C)-induced apoptosis
TNFSF10 plays an essential role in antiviral immunity-induced apoptosis, partly through type I IFN signaling
This dual role positions TNFSF10 at the intersection of tumor cell apoptosis and antiviral immunity, potentially explaining why its expression correlates with CD274 (PD-L1), a therapeutic target in cancer immunotherapy .
Researchers should design experiments that specifically distinguish between direct apoptotic effects and indirect immune-mediated effects when studying TNFSF10 function in complex biological systems.
Cancer cells employ multiple mechanisms to evade TNFSF10-induced apoptosis:
Resistance mechanisms and experimental countermeasures:
Research demonstrates that protective autophagy is activated in response to TNFSF10 treatment, as evidenced by:
Conversion of MAP1LC3B-I to MAP1LC3B-II
Reduction of SQSTM1/p62 expression levels
Further elevation of MAP1LC3B-II when combining TNFSF10 with chloroquine
To overcome resistance experimentally:
Perform combination treatments with sensitizing agents
Target multiple resistance mechanisms simultaneously
Use genetic approaches (CRISPR/siRNA) to identify specific resistance factors
Consider the tumor microenvironment context, as TNFSF10 affects immune cell infiltration
When designing sensitization experiments, include appropriate controls to distinguish between enhanced TRAIL receptor signaling versus alternative apoptotic mechanisms.
Genetic variations in TNFSF10 significantly impact its expression and function:
SNP rs13074711 in TNBC:
Located in a region with enhancer marks and open chromatin structure
Regulates TNFSF10 expression in triple-negative breast cancer (TNBC) cells
Expression correlates with SNP genotype:
SNP modulates c-MYB binding activity at the enhancer motif
Association with racial disparities:
TNFSF10 expression is consistently lower in African American (AA) cancer patients compared to European American (EA) patients
Statistically significant lower expression observed in colon and kidney cancers of AA patients
Potentially contributes to resistance to TNFSF10-driven apoptosis of cancer cells in AA patients
Other functional SNPs:
Three SNPs within the TNFSF10 gene have been associated with T4 effector memory lymphocyte radiosensitivity :
SNP | Chromosome Location | Association p-value |
---|---|---|
rs3815496 | Chr3:172,227,199 | 0.03 |
rs1131532 | Chr3:172,224,303 | 0.04 |
rs1131535 | Chr3:172,224,075 | 0.05 |
These same SNPs have been linked to risk and/or treatment outcomes in ovarian and breast cancer .
For experimental investigation of TNFSF10 genetic variants:
Use site-directed mutagenesis to recreate variants in expression systems
Employ CRISPR-Cas9 to introduce specific SNPs into model cell lines
Perform allele-specific expression analyses in heterozygous samples
Conduct reporter assays with wildtype versus variant enhancer sequences
TNFSF10's dual functionality presents unique opportunities for cancer immunotherapy:
Apoptotic and immune functions:
Direct tumor cell killing through death receptor signaling
Modulation of tumor microenvironment and immune cell infiltration
Correlation with expression of CD274 (PD-L1), suggesting potential synergy with checkpoint inhibitors
Therapeutic strategies leveraging TNFSF10:
Combination with immune checkpoint inhibitors:
Enhancement of antiviral immune signaling:
TNFSF10-based combination therapies:
Research models for studying these approaches should incorporate:
Immunocompetent animal models (avoid athymic nude or NOD/SCID mice)
Patient-derived xenografts in humanized mouse models
Ex vivo tumor slice cultures that preserve tumor microenvironment
Multi-parametric analysis of immune infiltrates following treatment
The 4T1 syngeneic mouse model has been successfully used to determine effects of TNFSF10-knockout on T-cell infiltration, demonstrating TNFSF10's role in regulating immune cell infiltration in the tumor microenvironment .
To comprehensively evaluate TNFSF10's impact on the tumor microenvironment and immune infiltration:
Experimental approaches:
Immunocompetent mouse models:
Multi-parameter flow cytometry:
Analyze tumor-infiltrating lymphocytes (TILs) including:
CD4+ T cells (helper, regulatory)
CD8+ T cells (cytotoxic)
NK cells
Myeloid populations (MDSCs, TAMs)
Include functional markers (activation, exhaustion, cytotoxicity)
Spatial analysis:
Multiplex immunohistochemistry to preserve spatial relationships
Quantify immune cell density and distribution
Assess proximity of immune cells to tumor cells
Digital spatial profiling for high-dimensional analysis
Transcriptomic approaches:
Bulk RNA-seq to identify pathway activation
Single-cell RNA-seq to define cellular heterogeneity
Correlate TNFSF10 expression with immune signatures
Key findings from research:
TNFSF10-knockout significantly reduced tumor-infiltrating CD4+ T cells compared to wild-type tumors
Expression of TNFSF10 correlates with antiviral gene expression rather than inflammatory or apoptosis-related genes
Expression correlates with CD274 (PD-L1), suggesting potential impact on checkpoint inhibitor response
When designing such studies, researchers should:
Include time-course analyses to capture dynamic changes
Compare multiple tumor models with varying baseline immune infiltration
Consider both membrane-bound and soluble forms of TNFSF10
Incorporate relevant genetic variations (e.g., rs13074711) that affect TNFSF10 expression
The choice of expression system significantly impacts recombinant TNFSF10 quality and activity:
Comparison of expression systems:
Expression System | Advantages | Limitations | Best Applications |
---|---|---|---|
E. coli | High yield; Cost-effective; Simpler purification | No glycosylation; Potential endotoxin; Refolding often required | Basic apoptosis assays; Structural studies; High-throughput screens |
HEK-293 cells | Human-like post-translational modifications; Proper folding; Lower endotoxin | Lower yield; Higher cost; More complex purification | Complex functional studies; In vivo applications; Clinical-grade protein production |
Wheat germ | Cell-free system; Reduces toxicity issues | Moderate yield; Less commonly used | Applications requiring reduced endotoxin without mammalian glycosylation |
Critical quality attributes to assess:
Oligomeric state: Native TNFSF10 functions as a homotrimer; recombinant protein should maintain this state
Endotoxin levels: Must be <0.2 EU/μg for cell culture applications, determined by LAL method
Specific activity: ED50 < 40 ng/mL in RPMI-8226 cell growth inhibition assay, corresponding to >2.5 × 10^4 units/mg
Purity: >95% as determined by SDS-PAGE and HPLC/SEC analysis
Distinguishing between membrane-bound TNFSF10 (mTRAIL) and soluble TNFSF10 (sTRAIL) presents specific technical challenges that researchers must address:
Methodological approaches to differentiate forms:
Flow cytometry:
Non-permeabilized cells detect only membrane-bound form
Comparison of permeabilized versus non-permeabilized samples differentiates total versus surface TNFSF10
Use anti-TNFSF10 antibodies that recognize extracellular domain
ELISA:
For culture supernatants/biological fluids to quantify sTRAIL
Important to generate standard curves with recombinant protein matching the form being measured
Western blot:
Different molecular weights: full-length (~32-33 kDa) versus soluble (~24-28 kDa)
Membrane fractionation protocols to separate cellular compartments
Metalloprotease inhibition:
Research findings on form-specific functions:
mTRAIL but not sTRAIL regulates radiation-induced apoptosis of T4EM lymphocytes
Adding recombinant human soluble TRAIL (rh-sTRAIL) has inhibiting effect on radiation-induced apoptosis
Higher cell concentrations reduce apoptosis, suggesting cell density affects mTRAIL-mediated effects
Experimental design considerations:
Include metalloprotease inhibitors to prevent shedding when studying mTRAIL
Account for cell density effects in functional assays
Use genetic approaches (mutation of cleavage sites) to create non-cleavable variants
Consider paracrine versus autocrine signaling in co-culture systems
Understanding the differential biology of membrane versus soluble TNFSF10 is particularly important given their potentially opposing effects in certain experimental contexts, as demonstrated in radiation-induced apoptosis studies .
Investigating TNFSF10's role in early-life respiratory infections and chronic lung disease requires specialized approaches:
Experimental models and challenges:
Neonatal mouse models:
Assessment parameters:
Histopathology: inflammation, mucus hypersecretion
Alveolar structure: morphometric analysis of alveolar enlargement
Pulmonary function: compliance, resistance measurements
Immunological markers: immune cell infiltration, cytokine profiling
Technical challenges:
Small sample size from neonatal specimens
Difficulty distinguishing developmental versus pathological changes
Need for age-appropriate controls at each timepoint
Long-term follow-up required to assess chronic effects
Key research findings:
TRAIL promotes infection-induced histopathology, inflammation, and mucus hypersecretion
Contributes to subsequent alveolar enlargement and impaired lung function
May offer therapeutic target for early-life respiratory infections and associated chronic lung disease
Methodological recommendations:
Use longitudinal study designs with matched controls
Employ both genetic (knockout) and pharmacological (neutralizing antibodies) approaches
Consider sex as a biological variable in analysis
Incorporate clinically relevant respiratory pathogens (Chlamydia, RSV)
Use micro-sampling techniques to maximize data from limited specimens
Correlate animal model findings with human clinical samples when available
This research area highlights the complex dual nature of TNFSF10 in both protection against infection and contribution to pathological processes, requiring careful experimental design to dissect these opposing functions.