TNFSF15 interacts with death receptor 3 (TNFRSF25/DR3) and decoy receptor 3 (TNFRSF6B/DcR3) to regulate immune responses and vascular homeostasis. Key mechanisms include:
TNF Homology Domain: Binds DR3 to activate NF-κB and MAPK pathways, promoting cytokine secretion (e.g., IL-1, IL-6) and apoptosis in endothelial cells .
Coiled-Coil Stalk: Enables trimerization for receptor cross-linking and signal amplification .
TNFSF15 is implicated in inflammatory and autoimmune diseases:
Inflammatory Bowel Disease (IBD): Elevated TNFSF15 levels correlate with Crohn’s disease and ulcerative colitis severity. A disease-risk polymorphism (rs4979462) increases TNFSF15 expression and intestinal inflammation .
Systemic Lupus Erythematosus (SLE): The rs4979462 variant is associated with serositis and thrombotic manifestations, suggesting a role in immune dysregulation .
Cancer: Anti-angiogenic effects via VEGF inhibition make TNFSF15 a candidate for tumor vasculature targeting .
Anti-Inflammatory Target: Neutralizing TNFSF15 may reduce DR3-mediated cytokine storms in autoimmune diseases .
Anti-Angiogenic Therapy: Inhibits endothelial cell proliferation, offering potential in oncology .
Factor | Impact | Source |
---|---|---|
Endotoxin Contamination | Requires <0.1 EU/µg for in vivo use | , |
Stability | Lyophilized storage at -20°C required | , |
Partial Activity | Lacks full-length receptor-binding capacity | , |
This Recombinant Human TNFSF15 protein offers significant potential for cancer research, providing insights into the role of this cytokine as a member of the tumor necrosis factor ligand superfamily. Known as TNF ligand-related molecule 1 (TL1) or vascular endothelial cell growth inhibitor (VEGI), TNFSF15 plays a crucial role in diverse biological processes, including apoptosis, immune regulation, and angiogenesis. Investigating the functions of TNFSF15 can offer valuable insights into cancer biology, potentially leading to the development of novel therapeutic strategies.
Produced using a precise E. coli expression system, our Tag-Free recombinant protein exhibits high purity (>97%, as determined by SDS-PAGE and HPLC) and minimal endotoxin contamination (<1.0 EU/μg). The protein corresponds to amino acids 72 to 251, encompassing a partial length of the TNFSF15 sequence. Maintaining its native structure, this protein ensures reliable and consistent results in your experiments.
Our recombinant TNFSF15 protein demonstrates full biological activity, effectively inducing apoptosis using human TF-1 cells. The ED50, determined through a dose-response assay, is less than 20 ng/ml, corresponding to a specific activity of > 5.0 × 10^4 IU/mg. Supplied as a lyophilized powder, this product offers exceptional stability and can be conveniently reconstituted for diverse experimental applications. Uncover the intricate mechanisms underlying cancer progression and angiogenesis with our high-quality TNFSF15 protein.
This protein serves as a receptor for TNFRSF25 and TNFRSF6B, mediating the activation of NF-kappa-B. It inhibits vascular endothelial growth and angiogenesis (in vitro) and promotes the activation of caspases and apoptosis.
Human TNFSF15, also known as Vascular Endothelial Growth Inhibitor (VEGI), is encoded by a gene located on chromosome 9q32 spanning approximately 18,850 bases . The mature TNFSF15 protein consists of 192 amino acids organized into three distinct functional domains: a TNF homology domain for receptor binding, a coiled-coil stalk region for trimerization, and an intracellular N-terminal region . It functions as a type II transmembrane protein with a single transmembrane domain, a small cytoplasmic domain, and an extracellular C-terminus . TNFSF15 acts as a cytokine that binds to TNFRSF25 (DR3) and the decoy receptor TNFRSF21/DR6, activating NF-kappaB and MAP kinases to regulate immune responses .
TNFSF15 demonstrates a distinct expression pattern across human tissues:
Predominantly expressed in endothelial cells
Notably absent in B and T lymphocytes
Gene expression varies by tissue type and can be affected by genetic polymorphisms
Expression levels fluctuate in disease states, showing reduction in certain asthma phenotypes and elevation in some autoimmune conditions
For experimental design considerations, researchers should account for tissue-specific expression patterns and inducible nature of TNFSF15 when selecting appropriate biological systems and stimulation conditions.
Upon binding to its receptors, TNFSF15 activates several key signaling pathways with cell type-specific outcomes:
In macrophages specifically, TNFSF15:DR3 interactions amplify pattern-recognition-receptor (PRR)-initiated signaling through TRADD/FADD/MALT-1 and caspase-8-mediated pathways, leading to autocrine IL-1 secretion . In contrast, in retinal cells, TNFSF15 inhibits GSDME-dependent pyroptosis by directly interacting with GSDME to prevent its cleavage by caspase-3 .
TNFSF15 interacts with two primary receptors that mediate distinct biological effects:
Death Receptor 3 (DR3/TNFRSF25): The main functional receptor predominantly expressed on activated T cells and macrophages. Binding to DR3 initiates downstream signaling cascades critical for immune responses . Through this interaction, TNFSF15 promotes T-cell activation, proliferation, and cytokine generation .
Decoy receptor TNFRSF21/DR6: Functions as a regulatory receptor that can bind TNFSF15 without initiating the full spectrum of downstream signaling .
For effective experimental analysis of TNFSF15 function, researchers should verify receptor expression in their cellular models using flow cytometry or western blotting. The interaction between TNFSF15 and DR3 is essential for T-cell immune responses in T-cell-mediated autoimmune diseases , while in macrophages, this interaction enhances PRR-initiated signaling and cytokine secretion .
For maximum experimental reproducibility and protein activity when working with recombinant TNFSF15:
Storage: Store lyophilized protein at -20°C; avoid repeated freeze/thaw cycles
Reconstitution: Reconstitute in sterile PBS to a concentration of 1 mg/mL
Working solutions: Prepare fresh dilutions in appropriate buffers for each experiment
Temperature considerations: Maintain at 4°C during short-term experimental procedures
Activity verification: Include functional assays to confirm protein activity in each experiment
Researchers should document handling procedures in detail to improve reproducibility across studies. When reconstituting the protein, allow complete dissolution through gentle rotation rather than vigorous vortexing to preserve the protein structure and function.
Selection of appropriate cell models depends on the specific TNFSF15 function under investigation:
When designing experiments, researchers should:
Verify receptor expression in selected cell lines before conducting experiments
Include appropriate positive and negative controls
Consider using primary cells when possible for greater physiological relevance
Account for potential species differences when translating findings from animal models
Several complementary methodological approaches can be employed to assess TNFSF15 activity:
Signaling pathway activation:
Cytokine production assessment:
ELISA or multiplex assays for secreted cytokines (IL-1, IL-6, TNF-α)
qRT-PCR for cytokine gene expression changes
Intracellular cytokine staining and flow cytometry
Functional assays:
Protein-protein interaction studies:
For comprehensive functional characterization, researchers should employ multiple complementary assays and include appropriate controls for each experimental approach.
Optimal TNFSF15 concentrations vary based on the specific biological readout:
Researchers should perform dose-response and time-course studies to determine optimal concentrations and time points for their specific experimental system. Additionally, comparing recombinant TNFSF15 effects to physiological levels observed in relevant disease contexts can provide important translational insights.
TNFSF15 has been identified as a significant risk factor in inflammatory bowel disease (IBD) through multiple mechanisms:
Genetic associations:
Mechanistic contributions:
Experimental approaches to investigate these associations include genotyping IBD cohorts for TNFSF15 variants, performing ex vivo studies with patient-derived cells, and using animal models with relevant TNFSF15 mutations or gene expression alterations.
TNFSF15 demonstrates complex roles in autoimmune disorders, with substantial evidence from systemic lupus erythematosus (SLE) research:
SLE associations:
The TNFSF15 rs4979462 gene variant significantly increases SLE risk in female subjects (OR = 2.6, 95% CI = 1.1–6.3)
The T-variant correlates with serositis and thrombotic manifestations
Serum TNFSF15 levels are elevated in SLE patients compared to healthy controls
TNFSF15 serum levels correlate with SLE disease activity (p = 0.012)
Immunomodulatory mechanisms:
These findings suggest TNFSF15 could serve as both a biomarker for disease activity and a potential therapeutic target in autoimmune disorders. Methodological approaches for further investigation include genotyping using PCR-RFLP with verification by direct sequencing, measurement of serum TNFSF15 using ELISA, and correlation analysis with clinical parameters.
Recent research has revealed a protective role for TNFSF15 in diabetic retinopathy (DR):
This protective mechanism represents a potential therapeutic avenue for diabetic retinopathy treatment through targeting the TNFSF15-GSDME interaction. Experimental approaches for investigating this pathway include high glucose culture models of retinal cells, assessment of pyroptosis markers, co-immunoprecipitation studies, and functional studies with TNFSF15 overexpression or knockdown.
Emerging evidence suggests TNFSF15 may play a role in asthma development through its effects on angiogenesis:
Genetic associations:
Proposed mechanism:
Gene interaction network:
These findings suggest that reduced TNFSF15 expression could promote asthma development through loss of angiogenesis inhibition. Future research directions include functional studies evaluating the effect of TNFSF15 on airway remodeling and investigating whether serum TNFSF15 levels could serve as a marker for persistent asthma development.
TNFSF15 plays a crucial role in amplifying pattern recognition receptor (PRR) responses through several mechanisms:
Enhancement of PRR signaling:
Molecular mechanism:
PRR types affected:
Disease relevance:
Research approaches to study this interaction include in vitro stimulation of macrophages with PRR ligands with and without TNFSF15, signal transduction analysis, and cytokine measurements using various assays.
As TNFSF15 is also known as Vascular Endothelial Growth Inhibitor (VEGI), several specialized techniques can assess its anti-angiogenic properties:
In vitro angiogenesis assays:
Endothelial cell proliferation assays
Tube formation assays on Matrigel
Wound healing/scratch assays
Transwell migration and invasion assays
Ex vivo techniques:
Aortic ring assays
Choroidal explant sprouting
Retinal explant culture
In vivo approaches:
Molecular methods:
Analysis of angiogenic/anti-angiogenic factor expression
VEGF signaling pathway component assessment
Endothelial cell gene expression profiling
Vessel density quantification in tissue samples
The anti-angiogenic properties of TNFSF15 appear particularly relevant in asthma pathophysiology, where reduced TNFSF15 levels might contribute to increased angiogenesis in asthmatic patients , and in diabetic retinopathy, where TNFSF15 provides protection against vascular damage .
Investigation of TNFSF15 polymorphisms requires comprehensive methodological approaches:
Genotyping strategies:
Functional validation:
Mechanism investigation:
For rs7856856 and asthma: Evidence suggests reduced TNFSF15 may promote angiogenesis
For rs6478108: Risk-carrier macrophages demonstrate increased TNFSF15 expression and PRR-induced signaling and cytokines
For rs4979462: Associated with increased SLE susceptibility and specific clinical manifestations
Clinical correlation approaches:
Case-control studies with appropriate population matching
Detailed phenotyping to identify variant-associated clinical features
Longitudinal studies to assess impact on disease progression
These genetic association studies provide valuable insights into disease mechanisms and potential personalized therapeutic approaches. When conducting such studies, researchers should consider population stratification, linkage disequilibrium with other variants, and tissue-specific effects.
The TNFSF15-DR3 signaling pathway presents several opportunities for therapeutic intervention:
Potential therapeutic strategies:
Disease-specific approaches:
Experimental validation methodologies:
Structure-based drug design targeting the TNFSF15-DR3 interface
Cell-based screening assays measuring NF-κB activation
Ex vivo testing in patient-derived cells
Animal models of specific diseases (colitis, asthma, retinopathy)
Therapeutic development targeting this pathway must consider tissue-specific effects, genetic variation that might affect response, and the balance between beneficial immune modulation and potential adverse effects.
Several apparent contradictions exist in TNFSF15 research literature that require careful interpretation:
Context-dependent effects:
Methodological approaches to resolve contradictions:
Tissue-specific and cell-type-specific studies to identify context-dependent responses
Time-course experiments to capture dynamic changes in TNFSF15 expression and function
Dose-response studies, as different concentrations may have opposing effects
Development of conditional knockout models to study tissue-specific roles
Research design considerations:
Use multiple experimental systems to validate findings
Include appropriate controls for each experimental approach
Consider genetic background effects in animal models and human studies
Account for environmental factors that may influence TNFSF15 expression
These contradictions likely reflect the complex, context-dependent biology of TNFSF15 rather than experimental inconsistencies. Researchers should consider the specific cellular environment, disease context, and genetic background when interpreting seemingly contradictory results.
Experimental design for combinatorial studies with TNFSF15 requires careful planning:
Interaction considerations:
Potential synergistic or antagonistic effects with other cytokines
Receptor competition or cross-regulation
Sequential versus simultaneous administration
Concentration ratios between TNFSF15 and other factors
Key combinations to consider:
Methodological approaches:
Factorial experimental designs to systematically test interactions
Response surface methodology to identify optimal combinations
Time-course studies with staggered administration
Single-cell analysis to identify responding subpopulations
Controls and analysis:
Include single-factor controls in parallel with combination treatments
Use appropriate statistical methods to detect interaction effects
Consider temporal aspects of signaling pathways
Document treatment sequences and concentrations precisely
These considerations are essential for understanding how TNFSF15 functions within the complex network of immune modulators in both physiological and pathological conditions.
Maintaining TNFSF15 stability is crucial for experimental reproducibility and reliable results:
Storage recommendations:
Reconstitution best practices:
Activity verification methods:
Include positive controls in functional assays
Verify protein integrity via SDS-PAGE
Confirm specific activity in relevant bioassays before critical experiments
Consider lot-to-lot variation when using commercial recombinant proteins
These stability considerations are essential for ensuring reproducible results across experiments and between different research groups studying TNFSF15 functions.
The distinct biological activities of membrane-bound versus soluble TNFSF15 require specialized approaches for differentiation:
Detection methods:
Flow cytometry with surface staining for membrane-bound TNFSF15
ELISA for soluble TNFSF15 in culture supernatants or biological fluids
Western blotting with appropriate antibodies recognizing different forms
Immunofluorescence microscopy to visualize cellular localization
Experimental approaches:
Functional discrimination:
Understanding the balance between membrane-bound and soluble TNFSF15 is particularly important in inflammatory conditions where TACE/ADAM17 activity may be altered, affecting the ratio between these forms and consequently modulating immune responses.