Recombinant Human Tumor necrosis factor ligand superfamily member 10 (TNFSF10), partial (Active)

Shipped with Ice Packs
In Stock

Description

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.

Product Specs

Buffer
0.2 µm filtered 40mM Tris-HCl, 300mM NaCl, 5% Trehalose, 5% Mannitol, 0.01% Tween80, 10% Glycerol, pH 7.0.
Description

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.

Form
Liquid or Lyophilized powder
Lead Time
Generally, we can ship the products within 1-3 working days after receiving your orders. Delivery times may vary depending on the purchase method or location. For specific delivery timelines, please consult your local distributors.
Please note: All our proteins are shipped with standard blue ice packs. If you require dry ice shipment, kindly inform us in advance, as additional fees may apply.
Shelf Life
The shelf life of our products is influenced by various factors, including storage conditions, buffer components, temperature, and the inherent stability of the protein.
Generally, liquid forms have a shelf life of 6 months at -20°C/-80°C. Lyophilized forms typically have a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. For multiple use, aliquot the product. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Apo 2 ligand; APO 2L; Apo-2 ligand; Apo-2L; APO2L; CD253; CD253 antigen; Chemokine tumor necrosis factor ligand superfamily member 10; Protein TRAIL; TL2; TNF Related Apoptosis Inducing Ligand; TNF related apoptosis inducing ligand TRAIL; TNF-related apoptosis-inducing ligand; TNF10_HUMAN; TNFSF10; TRAIL; Tumor necrosis factor (ligand) family member 10; Tumor Necrosis Factor (ligand) Superfamily Member 10; Tumor necrosis factor apoptosis inducing ligand splice variant delta; Tumor necrosis factor ligand superfamily member 10
Datasheet & Coa
Please contact us to get it.
Expression Region
115-281aa
Mol. Weight
19.5 kDa
Protein Length
Partial
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Cytokine that binds to TNFRSF10A/TRAILR1, TNFRSF10B/TRAILR2, TNFRSF10C/TRAILR3, TNFRSF10D/TRAILR4 and possibly also to TNFRSF11B/OPG. It induces apoptosis. Its activity might be modulated by binding to the decoy receptors TNFRSF10C/TRAILR3, TNFRSF10D/TRAILR4 and TNFRSF11B/OPG, which cannot induce apoptosis.
Gene References Into Functions
  1. TRAIL protects against insulin resistance, NAFLD, and vascular inflammation. Elevating TRAIL levels might be an attractive therapeutic approach to mitigate features of diabetes, as well as liver and vascular injury, commonly observed in individuals with NAFLD. PMID: 28507343
  2. Data strongly suggest that XIAP-mediated inhibition of final caspase-3 processing is the primary obstacle in TRAIL-induced apoptosis in NCI-H460 cells. This obstacle can be overcome by Smac in a Bcl-2 level-dependent manner. PMID: 29927992
  3. Patients exhibiting the homozygous AA genotype of TRAIL SNPs rs1131568 and rs1131579, and the TT genotype of the TRAIL SNP rs1131580, demonstrated lower overall survival and higher rates of extrahepatic recurrence (EHR) than patients with the wild type or heterozygous genotypes. PMID: 30051596
  4. TRAIL synergistically sensitized irradiation-induced apoptosis in glioblastoma stem-like cells by increasing DR5 expression and reducing cFLIP expression. PMID: 29777390
  5. Serum TRAIL concentrations are significantly reduced in patients with early preeclampsia. PMID: 29674187
  6. Combined treatment with ILz:rhTRAIL and bortezomib effectively induced cell death in both TRAIL-susceptible and TRAIL-resistant cancer cells through the intracellular TRAIL signaling pathway. The efficiency of cell death was dependent on the characteristics of TRAIL within the environment provided by bortezomib. The combination treatment-induced cell death was not regulated by bortezomib-induced ER stress response or by a PMID: 29661248
  7. A significant association was identified between TRAIL polymorphisms and the susceptibility and severity of intervertebral disc degeneration in Han Chinese. PMID: 29284457
  8. High expression levels of Galectin-3 and low expression levels of TRAIL were found to be positively correlated with shorter median survival time and overall survival time. PMID: 28925481
  9. Genes related to the extrinsic pathway of apoptosis, including a receptor for TRAIL. PMID: 30061204
  10. Nelfinavir induces ER stress in renal cancer cells, sensitizing them to TRAIL. PMID: 30061216
  11. The DR5-Cbl-b/c-Cbl-TRAF2 complex inhibited TRAIL-induced apoptosis by promoting TRAF2-mediated polyubiquitination of caspase-8 in gastric cancer cells. PMID: 28972304
  12. The combination of alpha-mangostin with TRAIL induced apoptosis of SAS cells through the mitochondrial pathway via activation of caspase-9 and -3/7, following release of cytochrome c. This apoptosis was induced by S/G2/M-phase arrest. Immunopositivity for c-Myc was observed in the cytoplasm of tumor cells in 16 (40%) of the 40 cases of human oral squamous cell carcinoma (HOSCC). PMID: 29039600
  13. This study characterized a distinct profile in juvenile systemic lupus erythematosus compared to adult SLE, comprising increased sFas, sTRAIL, and reduced sFasL, particularly in patients with active disease and nephritis. PMID: 28378099
  14. Notch1 activation also suppressed A549 cell apoptosis by inhibiting the PI3K/pAkt pathway and activating the caspase-3 pathway in conjunction with TRAIL. Combining Notch1 signaling with TRAIL inhibited PI3K, phosphorylated Akt, and phosphorylated STAT3 expressions. PMID: 28718726
  15. Our research indicates that executioner caspase activation of the apoptotic nuclease CAD/DFF40 is crucial for TRAIL-induced mutations in surviving cells. As exposure to chemotherapy drugs also activates apoptotic caspases and presumably CAD, we hypothesize that these pathways might also contribute to the mutagenesis induced by conventional chemotherapy drugs, potentially augmenting the mutations arising from direct DNA damage. PMID: 28981092
  16. Cultured HCN-2 neurons were incubated at various times with GITRL and/or TRAIL, followed by nucleic acid and protein expression measurements. HCN-2 cells do not express GITRL mRNA, but it is induced after treatment with TRAIL. Cells did not express the GITRL receptor GITR mRNA, neither in control cultures nor after treatment with TRAIL. TRAIL, when associated with GITRL, exerted additive toxic effects. PMID: 28524007
  17. DR5 plays a dual role in death and survival signaling, resulting in TRAIL resistance in cancer cells. PMID: 29048428
  18. Dynamin isoforms differentially regulate the endocytosis and apoptotic signaling downstream of TRAIL-death receptor (TRAIL-DR) complexes in cancer cells. TRAIL stimulation activates ryanodine receptor-mediated calcium release from endoplasmic reticulum stores, leading to calcineurin-mediated dephosphorylation and activation of Dyn1, TRAIL-DR endocytosis, and increased resistance to TRAIL-induced apoptosis. PMID: 28049841
  19. These findings suggest that the FasL -844T>C single nucleotide polymorphism is implicated in the susceptibility to hepatitis C virus in Egyptian patients. This is the first report involving TRAIL gene polymorphism in the risk of the disease. PMID: 28587866
  20. ONC201 exhibits potent antiproliferative and proapoptotic effects across a wide range of breast cancer subtypes, through TRAIL-dependent and TRAIL-independent mechanisms. The small-molecule ONC201 induces expression of TRAIL and its receptor DR5. ONC201 has entered clinical trials for advanced cancers. We demonstrate that ONC201 is efficacious against both triple-negative breast cancers (TNBC) and non-TNBC cells. PMID: 28424227
  21. Our data suggest that nanoparticle-engineered TRAIL-expressing human adipose-derived stem cells exhibit the therapeutically relevant behavior of "seek-and-destroy" tumortropic migration and could be a promising therapeutic approach to improve the treatment outcomes of patients with malignant brain tumors. PMID: 27849590
  22. CaP@LDL-mediated STAT3-decoy ODN delivery might be a promising new strategy for reversing TRAIL resistance in hepatocellular carcinoma therapy. PMID: 29158840
  23. The combination of RA/poly(I:C) synergizes to induce a bioactive autocrine/paracrine loop of type-I Interferons (IFNs) which is ultimately responsible for TRAIL and TRAIL-R1/2 expression upregulation, while inhibition of TRAIL-R3/4 expression is type-I IFN-independent. PMID: 28409399
  24. Our results demonstrated that nanovectorization of TRAIL with BNNTs enhanced its binding to both DR4 and DR5 receptors at 37 degrees C. Our novel nanovector could potentially be used for delivering TRAIL to cells for cancer treatment. PMID: 28120533
  25. In primary hyperparathyroidism, hyperplasias demonstrated the highest expression of TRAIL and Fas, whereas in adenomas it was increased compared to normal tissue, but lower than in hyperplasias. PMID: 27763797
  26. Long-term toxicity testing in cynomolgus monkeys did not reveal rAAV2-sTRAIL95-281-related toxic and side effects, except for the generation of anti-AAV and anti-sTRAIL antibodies. In conclusion, these data demonstrated that administration of rAAV2-sTRAIL95-281 in mice and cynomolgus monkeys is safe without obvious toxic and side effects to the animals. These findings provide insights into pharmacokinetics and safety for potential human clinical trials. PMID: 28429751
  27. We discovered that plumbagin could enhance TRAIL-induced apoptosis in Kasumi-1 cells. The mechanisms involve ROS-mediated upregulation of DR5 expression, caspase-8 activation, and inhibition of cFLIP expression. PMID: 28498435
  28. None of the crude extracts exhibited cytotoxic effects on normal mouse embryonic fibroblasts (MEF), with the exception of EGY34. Analysis of the signaling pathways underlying the sensitization of MDA-MB-231 cells to TRAIL-induced apoptosis, by western blotting, revealed that all crude extracts facilitated initiator caspase8/-10 activation upon TRAIL stimulation. PMID: 28440502
  29. This report identifies a novel mechanism for the TRAIL-induced apoptosis of TrkAIII-expressing NB cells, dependent on SHP/Src-mediated crosstalk between the TRAIL-receptor signaling pathway and TrkAIII. PMID: 27821809
  30. Our findings highlight a key role for c-Myc in TRAIL deregulation and as a biomarker of the anticancer action of HDACi in acute myeloid leukemia. PMID: 27358484
  31. Apoptosis induced by TRAIL is preferentially triggered by the TRAIL-R1 pathway in breast/lung tumor cells. PMID: 28039489
  32. These effects of metformin were accompanied by substantial reductions in the protein levels of XIAP, a negative regulator of TRAIL-induced apoptosis. Silencing XIAP in TNBC cells mimicked the TRAIL-sensitizing effects of metformin. Metformin also enhanced the antitumor effects of TRAIL in a metastatic murine TNBC model. PMID: 28324269
  33. siRNA silencing of CHOP significantly reduced cyproterone acetate-induced DR5 up-regulation and TRAIL sensitivity in prostate cancer cells. Our study reveals a novel effect of cyproterone acetate on apoptosis pathways in prostate cancer cells and raises the possibility that a combination of TRAIL with cyproterone acetate could be a promising strategy for treating castration-resistant prostate cancer. PMID: 28270124
  34. TRAIL-armed exosomes can induce apoptosis in cancer cells. PMID: 26944067
  35. TRAIL expression in CTLs was negatively correlated with the expression of perforin and granzyme B, and negatively correlated with CTLs apoptosis in severe aplastic anemia patients. PMID: 28631177
  36. Results provide convincing evidence that employing TRAIL combined with cisplatin/LA-12 could contribute to more effective killing of prostate cancer cells compared to the individual action of the drugs. This research offers new mechanistic insights into their cooperative anticancer action. PMID: 29182622
  37. UMI-77 enhances TRAIL-induced apoptosis by unsequestering Bim and Bak, providing a novel therapeutic strategy for the treatment of gliomas. PMID: 28337703
  38. These results reveal KDM4A as a key epigenetic silencer of TRAIL and DR5 in tumors. PMID: 27612013
  39. Low Trail expression is associated with esophageal cancer. PMID: 29055676
  40. Results demonstrate that TNF-Related Apoptosis Inducing Ligand (TRAIL/TNFSF10) mRNA level correlates with T4 effector-memory lymphocytes (T4EM) lymphocytes radiosensitivity. PMID: 26982083
  41. An imbalance in the apoptosis pathway, with dysregulation of p73 and TRAIL, appears to play a role in the oncogenesis of odontogenic tumors. PMID: 28025428
  42. Hepatitis B virus (HBV) X protein (HBx) restricts TNFSF10 receptor signaling through macroautophagy/autophagy-mediated degradation of TNFRSF10B/DR5, a TNFSF10 death receptor. PMID: 27740879
  43. Findings indicate that loss of function of the human TRAIL promoter due to the SNP mutation leads to reduced expression and impaired inducibility of TRAIL, resulting in enhanced growth and migration of tumor cells and ultimately contributing to the progression of hepatocellular carcinoma. PMID: 27580702
  44. These data show increased TRAIL mRNA expression in the airways and parenchyma in chronic obstructive pulmonary disease. PMID: 26555706
  45. TRAIL expression is associated with thyroid hormone levels in vivo. Additionally, TRAIL expression in peripheral blood mononuclear cells is stimulated by thyroid hormones in vitro. PMID: 28551332
  46. Antagonistic antibodies against the death receptors demonstrated that Apo2L/TRAIL mediated its apoptotic signals through activation of the TRAIL-R2 in PTHrP-expressing breast cancer cells. PMID: 23822995
  47. Co-treatment of carboplatin and autophagy inhibitor chloroquine increased lung tissue infiltration by CD4(+), FoxP3(+) lymphocytes and antigen-specific immune activation. Subsequent ex vivo experiments revealed the activation of carboplatin-related TRAIL-dependent apoptosis through caspase 8, along with a synergistic role of miR-155 in lung tissue infiltration by CD4(+), and FoxP3(+) lymphocytes. PMID: 27692344
  48. Caspase-8 can play two distinct roles in response to TRAIL receptor engagement: as a scaffold for assembling a Caspase-8-FADD-RIPK1 "FADDosome" complex, leading to NFkappaB-dependent inflammation, or as a protease that promotes apoptosis. PMID: 28212752
  49. Endogenous TRAIL/TRAIL-R-mediated CCL2 secretion promotes accumulation of tumor-supportive immune cells in the cancer microenvironment. PMID: 28212753
  50. BAY61-3606 sensitizes colon cancer cells to TRAIL-induced apoptosis by upregulating DR4 expression in a p53-dependent manner and inhibiting NF-kappaB activity. PMID: 27721019

Show More

Hide All

Database Links

HGNC: 11925

OMIM: 603598

KEGG: hsa:8743

STRING: 9606.ENSP00000241261

UniGene: Hs.478275

Protein Families
Tumor necrosis factor family
Subcellular Location
Cell membrane; Single-pass type II membrane protein. Secreted.
Tissue Specificity
Widespread; most predominant in spleen, lung and prostate.

Q&A

What is TNFSF10 and what are its primary functions in human biology?

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

  • Modulating immune cell function and proliferation

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.

How should recombinant TNFSF10 be stored and reconstituted for optimal activity?

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 .

What are the key differences between full-length and partial recombinant TNFSF10?

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

  • Most commonly used form for biological activity studies

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.

How should TNFSF10-induced apoptosis assays be designed and what controls are necessary?

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

  • Mitochondrial membrane potential assessment

Data should be presented showing both dose-response and time-course relationships, with statistical analysis of at least three independent experiments.

What methods are most appropriate for studying TNFSF10 expression at the mRNA and protein levels?

For comprehensive analysis of TNFSF10 expression:

mRNA expression analysis:

  • qRT-PCR: Use TaqMan or SYBR Green assays with properly validated primers

    • For TaqMan: Hs00921974_m1 probe is validated for human TNFSF10

    • Normalize to validated reference genes (e.g., RNA18S, GAPDH, ACTB)

    • Calculate fold change using the ΔΔCT method

  • 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

    • Recommended antibodies: anti-TNFSF10 (Cell Signaling #3219, 1:300 dilution)

    • Use tubulin (Cell Signaling #2128) or β-actin (Abcam #ab8227) as loading controls

    • Employ fluorescent secondary antibodies for quantification

  • 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.

How does TNFSF10 contribute to antiviral immune responses, and how is this distinct from its apoptotic function?

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.

What mechanisms explain TNFSF10 resistance in cancer cells and how can they be experimentally overcome?

Cancer cells employ multiple mechanisms to evade TNFSF10-induced apoptosis:

Resistance mechanisms and experimental countermeasures:

Resistance MechanismMolecular BasisExperimental Approach to Overcome
Decoy receptor expressionUpregulation of TRAIL-R3/R4 competes for ligand binding without triggering apoptosissiRNA knockdown of decoy receptors; selective DR4/DR5 agonistic antibodies
Defective death receptor signalingMutations or downregulation of DR4/DR5; defects in DISC formationHDAC inhibitors to upregulate DR expression; IAP antagonists to enhance DISC formation
Anti-apoptotic protein overexpressionElevated levels of c-FLIP, Bcl-2, Bcl-xL, or IAPsBH3 mimetics (ABT-737); Smac mimetics; XIAP inhibitors
Pro-survival pathway activationConstitutive NF-κB, PI3K/Akt, or MAPK signalingPathway-specific inhibitors (e.g., BAY 11-7082 for NF-κB)
Protective autophagyAutophagy induction following TRAIL treatmentChloroquine or 3-methyladenine to inhibit autophagy

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.

How does genetic variation in TNFSF10 affect its expression and function, particularly in cancer?

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:

    • CC genotype: highest TNFSF10 expression

    • TT genotype: lowest TNFSF10 expression

    • TC genotype: intermediate expression

  • 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 :

SNPChromosome LocationAssociation p-value
rs3815496Chr3:172,227,1990.03
rs1131532Chr3:172,224,3030.04
rs1131535Chr3:172,224,0750.05

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

How can TNFSF10's dual role in apoptosis and immune modulation be leveraged for cancer immunotherapy?

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:

    • TNFSF10 influences tumor-infiltrating lymphocytes (TILs)

    • TNFSF10-knockout tumors showed significantly reduced CD4+ T-cell infiltration

    • Tumors with high TIL density respond better to immune checkpoint inhibitors

  • Enhancement of antiviral immune signaling:

    • Activation of type I IFN pathways increases TNFSF10 expression

    • Antiviral innate immune responses promote antitumor immunity

    • Poly(I:C) or similar TLR agonists could enhance TNFSF10-mediated effects

  • TNFSF10-based combination therapies:

    • Target both direct apoptosis and immune activation

    • Combine with sensitizing agents to overcome resistance mechanisms

    • Consider genetic background (e.g., rs13074711 genotype) for personalized approaches

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 .

What methodological approaches best assess TNFSF10's effects on tumor microenvironment and immune cell infiltration?

To comprehensively evaluate TNFSF10's impact on the tumor microenvironment and immune infiltration:

Experimental approaches:

  • Immunocompetent mouse models:

    • Use syngeneic models (e.g., 4T1 breast cancer model)

    • Create TNFSF10-knockout lines using CRISPR-Cas9

    • Compare wild-type vs. knockout tumors for immune infiltration

    • Essential to avoid immune-deficient models that cannot exhibit actual tumor microenvironment

  • 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

How do different expression systems affect the quality and activity of recombinant TNFSF10?

The choice of expression system significantly impacts recombinant TNFSF10 quality and activity:

Comparison of expression systems:

Expression SystemAdvantagesLimitationsBest Applications
E. coliHigh yield; Cost-effective; Simpler purificationNo glycosylation; Potential endotoxin; Refolding often requiredBasic apoptosis assays; Structural studies; High-throughput screens
HEK-293 cellsHuman-like post-translational modifications; Proper folding; Lower endotoxinLower yield; Higher cost; More complex purificationComplex functional studies; In vivo applications; Clinical-grade protein production
Wheat germCell-free system; Reduces toxicity issuesModerate yield; Less commonly usedApplications 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

What technical challenges exist in distinguishing membrane-bound versus soluble TNFSF10 in experimental systems?

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:

    • 1,10-phenanthroline treatment blocks TNFSF10 shedding

    • Increased level of mTRAIL after treatment indicates active shedding

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 .

How can researchers address the challenges of investigating TNFSF10 in early-life respiratory infections and subsequent chronic lung disease?

Investigating TNFSF10's role in early-life respiratory infections and chronic lung disease requires specialized approaches:

Experimental models and challenges:

  • Neonatal mouse models:

    • TRAIL-deficient neonatal mice reveal TRAIL's role in infection-induced pathology

    • Neutralizing antibodies against TRAIL and its receptors in wild-type mice provide comparative data

    • Critical timing considerations for infection and analysis (developmental windows)

  • 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.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.