Recombinant Human Tumor necrosis factor receptor superfamily member 10B (TNFRSF10B), partial (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity and Functional Validation

The recombinant protein demonstrates full biological activity by neutralizing endogenous TRAIL, as evidenced by its ability to suppress TRAIL-induced TNF production in lipopolysaccharide (LPS)-stimulated human peripheral blood mononuclear cells (PBMCs) . Key functional assays include:

  • Apoptosis Inhibition: Binds TRAIL with high affinity (ED50: 0.75–6 ng/mL) , blocking its interaction with endogenous receptors and preventing caspase-8 activation .

  • NF-κB Modulation: Participates in ER stress-induced apoptosis pathways .

  • Therapeutic Synergy: Enhances cytotoxicity when combined with agents like IFN-β or cyproterone acetate in prostate and glioma cancer models .

Production and Quality Control

The protein is produced via recombinant DNA technology, with rigorous quality controls:

  • Expression: Optimized in E. coli for cost-effectiveness or mammalian cells (HEK-293T) for post-translational modifications .

  • Purification: Affinity chromatography (e.g., Strep-Tactin columns) ensures high purity .

  • Stability: Lyophilized powder remains stable for 12 months at -70°C; reconstituted aliquots are stable for 3 months at -20°C .

Cancer Biology

  • TRAIL Resistance Mechanisms: Used to study dysregulated TRAIL-R2 expression in breast, colorectal, and glioblastoma cancers .

  • Therapeutic Targeting: Demonstrates synergy with chemotherapy and radiation by upregulating DR5 .

Immunology

  • Inflammation Modulation: Suppresses LPS-induced TNF production in PBMCs, highlighting its role in immune regulation .

Key Research Findings

StudyKey InsightCitation
TRAIL-R2/DR5 upregulation under ER stressEssential for stress-induced apoptosis in glutamine-addicted tumors
IFN-β sensitizes glioma cells to TRAILDR5 upregulation enhances apoptosis via NF-κB inactivation
Cyproterone acetate synergizes with TRAIL in prostate cancerNovel combination therapy for castration-resistant cancer
SKR compound cytotoxicity mechanismInvolves TRAIL-R2-mediated apoptosis in hypoxic cancer cells

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 20 mM phosphate buffer (PB), 150 mM sodium chloride (NaCl), pH 7.4.
Form
Lyophilized powder
Lead Time
Typically, we can ship the products within 5-10 business days after receiving your order. Delivery time may vary depending on the shipping method and location. Please consult your local distributor for specific delivery times.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents are at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer ingredients, storage temperature, and the stability of the protein itself. Generally, liquid forms have a shelf life of 6 months at -20°C/-80°C, while lyophilized forms have a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal 6xHis-hFc-tagged
Synonyms
TNFRSF10B; DR5; KILLER; TRAILR2; TRICK2; ZTNFR9; UNQ160/PRO186; Tumor necrosis factor receptor superfamily member 10B; Death receptor 5; TNF-related apoptosis-inducing ligand receptor 2; TRAIL receptor 2; TRAIL-R2; CD antigen CD262
Datasheet & Coa
Please contact us to get it.
Expression Region
56-182aa
Mol. Weight
42.2 kDa
Protein Length
Partial
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Cancer
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
This protein is a receptor for the cytotoxic ligand TNFSF10/TRAIL. The adapter molecule FADD recruits caspase-8 to the activated receptor. The resulting death-inducing signaling complex (DISC) activates caspase-8 proteolytically, which initiates the subsequent cascade of caspases (aspartate-specific cysteine proteases) mediating apoptosis. It promotes the activation of NF-kappa-B and is essential for ER stress-induced apoptosis.
Gene References Into Functions
  1. In vivo studies confirmed that the anti-tumor activity of bigelovin in colorectal cancer (CRC) was mediated through the induction of apoptosis by upregulating DR5 and increasing ROS. These results strongly suggest that bigelovin has potential as a therapeutic agent for CRC patients. PMID: 28181527
  2. GDF-15 and TRAIL-R2 were identified as the most powerful Proximity Extension Assay chip biomarkers for predicting long-term all-cause mortality in patients with acute myocardial infarction. PMID: 28762762
  3. In contrast to apoptosis, necroptotic signaling was activated similarly by both DR4- or DR5-specific ligands. This study provides the first systematic insight into DR4-/DR5-specific signaling in colorectal and pancreatic cancer cells. PMID: 29278689
  4. These data suggest that humanized anti-TRAIL-R2 monoclonal antibodies or second-generation antibodies may have significant clinical utility in cancer immunotherapy. PMID: 28748573
  5. Pharmacological application of Golgi stress leads to the induction of death receptors (DRs) 4 and 5. DR4 appears to be primarily responsible for initiating cell death downstream of Golgi stress, while DR5 seems to be more important for cell death triggered by endoplasmic reticulum (ER) stress in specific cancer cell lines. PMID: 28981087
  6. Knocking-down the TRAIL-DR5 gene in breast cancer cells MCF-7 significantly decreased the mRNA and protein levels of autophagy-related factors. PMID: 29268854
  7. Antineoplastic agents etoposide (ET) and doxorubicin enhance the expression of Death receptor 5 (DR5) in triple-negative breast cancer (TNBC) cells. DR5 residue SerB68 is crucial in mediating the receptor-drug interaction. Apoptosis and DR5 expression are induced in xenograft mice and in TNBC patient-derived metastatic cells after treatment with TNF-Related Apoptosis-Inducing Ligand (TRAIL) and ET. PMID: 28702823
  8. DR5, BIRC5/Survivin, XIAP, c-IAP1, and c-IAP2 mRNA expression are significantly deregulated in CRC and could provide a panel of markers with significant discriminatory value between CRC and normal colorectal tissue. PMID: 27827395
  9. The B-Raf inhibitor PLX4032 induces DR5 upregulation exclusively in Ras-mutant cancer cells; this effect is dependent on Ras/c-Raf/MEK/ERK signaling activation. PMID: 27222248
  10. EPHB6 induces marked fragmentation of the mitochondrial network in breast cancer cells of triple-negative origin. This response renders cancer cells more susceptible to DR5-mediated apoptosis. PMID: 27788485
  11. Both S1P and caspase-8 are critical for TRAF2 stabilization, polyubiquitination, subsequent activation of JNK/AP1 signaling and MMP1 expression, and final promotion of cell invasion. PMID: 28482915
  12. DNA fragmentation, mitochondrial membrane potential, and western blot analyses showed that MIC inhibited the growth of these cells by both mitochondrial-mediated and death receptor (DR5)-mediated apoptosis pathways. PMID: 28498480
  13. Targeting of lysosomes by chloroquine deregulates DR5 trafficking and abrogates 5-FU- but not TRAIL-stimulated cell elimination, suggesting a novel mechanism for receptor activation. PMID: 27506940
  14. siRNA silencing of CHOP significantly reduced cyproterone acetate-induced DR5 up-regulation and TRAIL sensitivity in prostate cancer cells. This study reveals a novel effect of cyproterone acetate on apoptosis pathways in prostate cancer cells and suggests that a combination of TRAIL with cyproterone acetate could be a promising strategy for treating castration-resistant prostate cancer. PMID: 28270124
  15. PU.1 supports TRAIL-induced cell death by inhibiting RelA-mediated cell survival and inducing DR5 expression. PMID: 28362429
  16. The results demonstrated CaM binding to DR5-mediated DISC in a calcium-dependent manner and may identify CaM as a key regulator of DR5-mediated DISC formation for apoptosis in breast cancer. PMID: 28092099
  17. The oncogene-like extracellular miR-1246 could act as a signaling messenger between irradiated and non-irradiated lung cancer cells, and more importantly, it contributes to cell radioresistance by directly suppressing the DR5 gene. PMID: 27129166
  18. Data show that the 4EGI-1 compound induced apoptosis in nasopharyngeal carcinoma cells through the death receptor 5 (DR5) on 4E-BP1 dephosphorylation, exerting a positive influence on their anti-tumor activities. PMID: 26942880
  19. Results show that downregulation of DR4 and DR5 by SLC26A2 confers resistance to TRAIL. PMID: 28108622
  20. This study provides direct biophysical evidence that Death Receptor 5 disulfide-linked transmembrane (TM)-dimers open in response to ligand binding. Then, to probe the importance of the closed-to-open TM domain transition in the overall energetics of receptor activation, point-mutants (alanine to phenylalanine) in the predicted, tightly packed TM domain dimer interface were designed and tested. PMID: 28746849
  21. Oridonin analog CYD-6-28 induces apoptosis at least partially by inducing the expression of death receptor 5 in breast neoplasms. PMID: 27387452
  22. The authors show that cholesterol is necessary for the covalent dimerization of DR5 transmembrane domains. PMID: 27720987
  23. Mono treatment with lexatumumab was not sufficient to induce apoptosis in pancreatic cancer cells, whereas focal adhesion kinase inhibitor PF573228 significantly sensitized lexatumumab-induced apoptosis. Western blotting analysis revealed that lexatumumab and PF573228 combination treatment increased death receptor 5 but decreased Bcl-xL expression. PMID: 28459212
  24. MG132 possesses anti-gallbladder cancer potential that correlates with regulation of the DR5-dependent pathway. PMID: 27277541
  25. CAPE/TRAIL stimulated apoptosis through the binding of TRAIL to DR5. Moreover, expression of transcription factor C/EBP homologous protein (CHOP) markedly increased in response to CAPE, and transient knockdown of CHOP abolished CAPE/TRAIL-mediated apoptosis. PMID: 27260301
  26. Decreased levels of placental TRAIL-R2 and previous C-section were found to be significantly correlated to placenta accreta. PMID: 26992667
  27. Results show that calmodulin (CaM) directly binds to death receptor-5 (DR5) in a calcium-dependent manner in breast cancer cells. PMID: 27129269
  28. Bee venom inhibits colon cancer cell growth, and these anti-proliferative effects may be related to the induction of apoptosis by activation of DR4 and DR5 and inhibition of NF-kappaB activity. PMID: 26561202
  29. A study involving a relatively large sample size showed that TNFRSF10 eQTL SNPs within lncRNAs might influence both hepatocellular carcinoma development and HBV infection. PMID: 26297860
  30. Data show that when death receptor 5 (DR5) is suppressed, caspase-8 may recruit and stabilize TNF receptor-associated factor 2 (TRAF2) to form a metastasis and invasion signaling complex, resulting in activation of ERK signaling. PMID: 26510914
  31. Synthetic lipid bilayers displaying the membrane protein ligand Apo2L/TRAIL were used to stimulate death receptor-expressing cells in a modular, scalable format. PMID: 26458551
  32. Methionine Deprivation Induces a Targetable Vulnerability in Triple-Negative Breast Cancer Cells by Enhancing TRAIL Receptor-2 Expression. PMID: 25724522
  33. This study investigated an array of TRAIL-R1 and TRAIL-R2 specific variants on pancreatic cancer cells. PMID: 26138346
  34. The expression of two proapoptotic genes, FAS and DR5, was significantly lower in tumor samples than in adjacent normal tissues. PMID: 25795228
  35. In an osteotropic variant of MDA-MB-231 breast cancer cells, TRAIL-R2 knockdown leads to downregulation of HMGA2, p-Src, p-Akt, and CXCR4 and increased E-cadherin expression. These changes diminished the occurrence of skeletal metastases in vivo. PMID: 25909161
  36. The mutant genotype (CT+TT) of DR5 (rs1047266) may exert a negative synergistic effect on Crohn's disease. PMID: 26418999
  37. This study investigated the apoptosis of hepatic stellate cells induced by SEA; it was found that apoptosis could be reduced in hepatic stellate cells treated with p53-specific siRNA and in hepatic stellate cells treated with DR5-specific shRNA. PMID: 25144704
  38. These findings strongly suggest that FLLL12 induces apoptosis of lung cancer cell lines by posttranscriptional regulation of DR5 through activation of protein tyrosine phosphatase(s). PMID: 25917567
  39. These findings highlight novel mechanisms underlying endoplasmic reticulum stress-induced TNFRSF10A and TNFRSF10B expressions and apoptosis. PMID: 25770212
  40. DR5 expression is dramatically reduced as a function of higher prostate tumor grade. PMID: 25174820
  41. There was a statistically significant association between DR5 expression and the tumor site of basal cell and squamous cell carcinoma skin cancers. PMID: 24212133
  42. Data suggest that H-Ras inhibits TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through downregulation of surface death receptors DR4/DR5. PMID: 25026275
  43. Over-expression of TRAIL-R2 is associated with breast cancer. PMID: 25230899
  44. The results show that both TRAIL-R1 and TRAIL-R2 are highly expressed on human oligodendrocyte progenitors. PMID: 25845236
  45. Further analysis demonstrated that PARP inhibitor treatment results in activation of the FAS and TNFRSF10B (death receptor 5 (DR5)) promoters, increased Fas and DR5 mRNA, and elevated cell surface expression of these receptors in sensitized cells. PMID: 24895135
  46. This study demonstrated lower apoptosis correlated with a deficiency of DR5 cell surface expression by CD4 T cells upon HIV-1 stimulation. PMID: 25110157
  47. This report highlights RR5 up-regulation in alveolar epithelial cells from idiopathic pulmonary fibrosis patients. PMID: 24551275
  48. Primary EOC is associated with lower TRAIL-R2 and BCL2 expression levels, while metastatic EOC is associated with higher expression of these genes. PMID: 24190693
  49. Parthenolide triggers extrinsic apoptosis by up-regulating TNFRSF10B and intrinsic apoptosis through increasing the expression of PMAIP1. PMID: 24387758
  50. Cotreatment with MESC and an ERK inhibitor (PD98059) significantly increased the expression of DR5 to induce apoptosis. This suggests that MESC may induce apoptosis via the ERK pathway and may be a potential anticancer drug candidate against human oral MEC. PMID: 24270523

Show More

Hide All

Database Links

HGNC: 11905

OMIM: 275355

KEGG: hsa:8795

STRING: 9606.ENSP00000276431

UniGene: Hs.521456

Involvement In Disease
Squamous cell carcinoma of the head and neck (HNSCC)
Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Widely expressed in adult and fetal tissues; very highly expressed in tumor cell lines such as HeLaS3, K-562, HL-60, SW480, A-549 and G-361; highly expressed in heart, peripheral blood lymphocytes, liver, pancreas, spleen, thymus, prostate, ovary, uterus,

Q&A

What is TNFRSF10B and what are its primary functions in cellular systems?

TNFRSF10B (also known as DR5, TRAIL-R2, CD262, KILLER, TRICK2) is a type 1 membrane protein belonging to the TNF receptor superfamily. It contains extracellular cysteine-rich domains, a transmembrane domain, and a cytoplasmic death domain . The protein is located at chromosome 8p21.3 and is one of five known receptors for TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) .

TNFRSF10B primarily functions as a death receptor that, upon binding trimeric TRAIL, triggers apoptosis through the extrinsic death pathway. When activated, it forms a death-inducing signaling complex (DISC) containing pro-caspase-8, ultimately leading to programmed cell death . Most notably, TRAIL specifically targets fully transformed cells without significantly affecting normal ones, making TNFRSF10B a promising target for cancer therapeutics .

How does TNFRSF10B differ from other TRAIL receptors in the TNF receptor family?

  • TNFRSF10B contains a functional intracellular death domain capable of transducing apoptotic signals

  • The decoy receptors compete for TRAIL binding but cannot initiate apoptosis

  • Unlike some receptors, there are no known rat or mouse homologs to TNFRSF10B

The ratio of functional death receptors to decoy receptors often determines cellular sensitivity to TRAIL-induced apoptosis.

What genomic alterations of TNFRSF10B are commonly observed in cancer tissues?

Genomic and expression alterations of TNFRSF10B vary significantly across cancer types and subtypes:

Cancer TypeCommon TNFRSF10B AlterationsReference
HPV(-) HNSCCHigher percentage of one-copy loss
HPV(+) HNSCCMore neutral or one-copy gain
Oropharyngeal tumorsHigher frequency of one-copy gain
Laryngeal and oral cavity tumorsHigher frequency of one-copy deletions

In head and neck squamous cell carcinoma (HNSCC), the TRAIL receptor family members (TNFRSF10A/B/C/D) exhibit significant differences in copy number variations (CNV) between tumors with different HPV status . These genomic alterations may contribute to differential sensitivity to TRAIL-based therapies and agents targeting TRAILRs.

What are the optimal reconstitution and storage conditions for recombinant TNFRSF10B proteins?

For optimal activity of recombinant TNFRSF10B proteins, researchers should follow these guidelines:

Reconstitution:

  • Lyophilized forms should be reconstituted at 500 μg/mL in PBS

  • Allow complete solubilization before using in experiments

Storage:

  • Use manual defrost freezers and avoid repeated freeze-thaw cycles

  • Store reconstituted protein at temperatures recommended by manufacturers (typically -20°C to -80°C)

  • Ship at ambient temperature but store immediately upon receipt

These conditions are critical for maintaining protein stability and functional activity in experimental settings.

Why is crosslinking necessary for some recombinant TNFRSF10B formats?

Crosslinking is essential for His-tagged TNFRSF10B formats because:

  • TNFRSF10B requires receptor trimerization for optimal biological activity

  • In natural systems, trimeric TRAIL induces oligomerization of the receptor, which is necessary for DISC formation and downstream signaling

  • Recombinant forms may not spontaneously form the optimal configuration without additional crosslinking

The crosslinking antibody (e.g., anti-6x histidine) enhances biological activity by promoting receptor clustering similar to what occurs with endogenous TRAIL binding . Without proper crosslinking, the recombinant protein may not achieve the same level of specific activity described in product datasheets.

What bioassays are most appropriate for evaluating TNFRSF10B functional activity?

Several bioassays can effectively measure TNFRSF10B functional activity:

  • Cytotoxicity Inhibition Assay:

    • Recombinant TRAIL R2/TNFRSF10B can inhibit TRAIL-mediated cytotoxicity in L-929 mouse fibroblast cells

    • The ED50 for this inhibitory effect is typically 0.75-6 ng/mL

  • Apoptosis Detection:

    • Flow cytometry with Annexin V/PI staining to quantify early and late apoptotic cells

    • Caspase-8 and caspase-3 activation assays to measure downstream effector activation

    • PARP cleavage detection by Western blot

  • Receptor Binding Assays:

    • Competitive binding assays with labeled TRAIL to measure receptor-ligand interactions

    • Surface plasmon resonance to determine binding kinetics and affinity

When designing these assays, it's crucial to include appropriate positive and negative controls to ensure the specificity of TNFRSF10B-mediated effects.

How does the HPV status affect TNFRSF10B expression and function in head and neck cancers?

HPV status significantly influences TNFRSF10B genomic alterations and potentially its function in head and neck squamous cell carcinoma (HNSCC):

HPV(+) HNSCC:

  • More frequently display neutral or one-copy gain of TNFRSF10B

  • Harbor less frequent FADD amplification and overexpression

  • Show more frequent shallow deletion of BIRC2/3 loci

  • Are generally less sensitive to SMAC mimetic birinapant

HPV(-) HNSCC:

  • Have higher percentage of one-copy loss of TNFRSF10B

  • Show different patterns of copy number variations of death pathway molecules

These distinct genomic profiles suggest that HPV status may be an important consideration when developing TRAIL-based therapeutic strategies for HNSCC. Oropharyngeal tumors, which are often HPV(+), showed the highest percentage of one-copy loss of FADD and BIRC2/3, with more frequent gain in TRAIL and its receptors .

What molecular mechanisms link cellular senescence to TNFRSF10B expression?

Cellular senescence involves a biphasic program that affects TNFRSF10B expression:

  • Early Phase:

    • Activation of senescence-associated secretory (SAS) factors

    • Upregulation of inflammatory and anti-apoptogenic genes

  • Late Phase:

    • Repression of DNA and chromatin components

    • Downregulation of CFLAR (which encodes c-FLIP, an inhibitor of apoptosis)

    • Increased expression of TNFRSF10B/DR5

This temporal regulation creates a window of vulnerability where senescent cells or their neighboring premalignant cells may become sensitized to TRAIL-induced apoptosis. The senescence secretome contains factors that lead to activation of the TRAIL pathway in premalignant cells, suggesting potential therapeutic opportunities for intervention at benign tumor stages .

How do genomic co-alterations of TNFRSF10B and other death pathway molecules affect therapeutic responses?

The therapeutic response to TRAIL-based treatments is influenced by the complex interplay of genomic alterations across multiple death pathway molecules:

MoleculeCommon AlterationsPotential Impact on Therapy
FADDAmplification in HPV(-), deletion in HPV(+)Affects DISC formation and apoptotic signaling
BIRC2/3Gene amplification, deletion varies by HPV statusModulates sensitivity to SMAC mimetics
XIAPGene amplification, variable deletionMay confer resistance to apoptosis
TRAIL receptorsCo-deletion or co-amplificationAlters balance of death vs. decoy receptors

These co-alterations create distinct molecular profiles that can predict sensitivity to various therapeutic approaches. For example, tumors with TNFRSF10B gain but FADD loss might respond differently to TRAIL than tumors with both components intact. Similarly, tumors with BIRC2/3 amplification may require combination therapy with SMAC mimetics to overcome resistance .

What factors contribute to variable TRAIL sensitivity despite TNFRSF10B expression?

Several factors can explain discrepancies between TNFRSF10B expression and TRAIL sensitivity:

  • Receptor Dynamics:

    • Post-translational modifications affecting receptor function

    • Differential receptor localization in membrane vs. cytoplasm

    • Variations in receptor clustering efficiency

  • Downstream Pathway Status:

    • Expression levels of anti-apoptotic proteins (c-FLIP, IAPs, Bcl-2 family)

    • Caspase-8 activation efficiency

    • Presence of downstream inhibitors

  • Experimental Considerations:

    • TRAIL preparation quality and concentration

    • Duration of treatment

    • Cell density and culture conditions

When encountering unexpected resistance, researchers should systematically evaluate each of these factors to identify the specific mechanism operating in their model system.

How can researchers optimize experimental design when studying TNFRSF10B-mediated apoptosis?

A robust experimental design for studying TNFRSF10B-mediated apoptosis should include:

  • Appropriate Controls:

    • Positive control (known TRAIL-sensitive cell line)

    • Negative control (either TRAIL-resistant cell line or TRAIL + pan-caspase inhibitor)

    • TNFRSF10B blocking antibody control

  • Multiple Readouts:

    • Early apoptosis markers (phosphatidylserine externalization)

    • Executioner caspase activation (caspase-3/7)

    • Late apoptosis markers (DNA fragmentation)

  • Validation Approaches:

    • Confirm TNFRSF10B expression by flow cytometry and Western blot

    • Verify the absence of mutations in the death domain

    • Assess expression of other TRAIL receptors (TNFRSF10A/C/D)

  • Optimized Reagents:

    • Properly reconstituted and stored recombinant proteins

    • Validated crosslinking antibodies when using His-tagged formats

    • Fresh preparations of all reagents for each experiment

This comprehensive approach minimizes variability and increases confidence in experimental results.

How might TNFRSF10B-targeted therapeutics overcome resistance mechanisms in cancer?

Innovative approaches to overcome TRAIL resistance include:

  • Receptor-Specific Targeting:

    • DR5-selective TRAIL variants with enhanced affinity

    • DR5-B functionalized with tumor-penetrating peptides like iRGD

    • Antibody-drug conjugates specifically targeting TNFRSF10B

  • Combination Strategies:

    • Sensitization with agents that upregulate TNFRSF10B expression

    • Co-targeting anti-apoptotic proteins (IAPs, c-FLIP)

    • Exploiting senescence-induced vulnerability periods

  • Novel Delivery Systems:

    • Nanoparticle formulations to enhance delivery and stability

    • Multimeric TRAIL formats to promote more effective receptor clustering

    • Tumor microenvironment-responsive activation systems

These approaches take advantage of the tumor-selective nature of TRAIL-induced apoptosis while addressing common resistance mechanisms .

What is the significance of TNFRSF10B in non-apoptotic signaling pathways?

Beyond its canonical role in apoptosis, TNFRSF10B can activate several non-apoptotic pathways:

  • Inflammatory Signaling:

    • NF-κB activation leading to pro-inflammatory cytokine production

    • MAPK pathway activation affecting cell survival and proliferation

  • Cell Migration and Invasion:

    • Potential role in activating Src family kinases

    • Effects on matrix metalloproteinase expression

  • Metabolism:

    • Emerging evidence suggesting links to metabolic pathways

    • Potential crosstalk with glutamine utilization pathways

Understanding these non-canonical functions is essential for predicting potential off-target effects of TNFRSF10B-targeted therapies and may reveal new therapeutic opportunities.

How does the TNFRSF10B genomic landscape vary across different cancer types?

TNFRSF10B genomic alterations show distinct patterns across cancer types:

Cancer TypeAssociated TNFRSF10B AlterationsPublication Count
Breast CancerVarious alterations23
Lung CancerVarious alterations11
Colorectal CancerVarious alterations10
Prostate CancerVarious alterations7
Head and Neck CancersCopy number variations7

These cancer-specific patterns suggest that TNFRSF10B may play different roles in the pathogenesis of various malignancies . Researchers should consider cancer type-specific genomic contexts when designing experiments and interpreting results related to TNFRSF10B function.

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.