Recombinant Human Tumor necrosis factor ligand superfamily member 15 protein (TNFSF15), partial (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity and Mechanism of Action

TNFSF15 interacts with death receptor 3 (TNFRSF25/DR3) and decoy receptor 3 (TNFRSF6B/DcR3) to regulate immune responses and vascular homeostasis. Key mechanisms include:

Functional Domains and Signaling Pathways

  • 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 .

Key Biological Effects

FunctionMechanismOutcomeSource
Immune ModulationDR3-dependent NF-κB/MAPK activationT-cell proliferation, cytokine release ,
Anti-AngiogenesisVEGF suppression via MAPK/NF-κB inhibitionReduced endothelial cell growth ,
Gut Barrier MaintenanceModulation of intestinal immune responseProtection against dysbiosis ,

Disease-Associated Studies

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 .

Experimental Models

ModelObservationSource
bEnd.3 Endothelial CellsTNFSF15 treatment ↓ VEGF mRNA/protein by 2–3×
Human MDMTNFSF15 amplifies NOD2-induced cytokines
Mouse Intestinal Myeloid CellsTNFSF15 enhances bacterial-induced cytokines

Advantages

  • 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 .

Challenges

FactorImpactSource
Endotoxin ContaminationRequires <0.1 EU/µg for in vivo use ,
StabilityLyophilized storage at -20°C required ,
Partial ActivityLacks full-length receptor-binding capacity ,

Product Comparisons

ProductHostTagApplicationSource
RPC28810E. coliTag-freeApoptosis assays
TNFSF15-1071HE. coliNoneBioactivity testing
TNFSF15-150HHEK293His-FlagReceptor-binding studies

Product Specs

Buffer
0.2 μm filtered PBS, pH 7.4, with 0.02% Tween-20, lyophilized
Description

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.

Form
Liquid or Lyophilized powder
Lead Time
5-10 business days
Shelf Life
The shelf life is influenced by various factors, including storage state, buffer ingredients, storage temperature, and the protein's inherent stability. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
TNFSF15; TL1; VEGI; Tumor necrosis factor ligand superfamily member 15; TNF ligand-related molecule 1; Vascular endothelial cell growth inhibitor
Datasheet & Coa
Please contact us to get it.
Expression Region
72-251aa
Mol. Weight
20.5 kDa
Protein Length
Partial
Purity
>97% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function

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.

Gene References Into Functions
  1. TNFSF15 polymorphisms may contribute to genetic susceptibility of inflammatory bowel disease (Meta-Analysis) PMID: 29873318
  2. TL1A modulated Rheumatoid arthritis-fibroblast-like synoviocytes migration and Indian hedgehog signaling pathway using TNFR2. PMID: 29748156
  3. TL1A can induce tumor cell proliferation and promote the occurrence of colitis-associated colorectal cancer by activating Wnt/beta-catenin pathway. PMID: 29796912
  4. TNFSF15, a cytokine mainly produced by blood endothelial cells, facilitates tumor lymphangiogenesis by upregulating VEGFC expression in A549 cells. PMID: 29890027
  5. Results suggested that TNFSF15 (rs3810936 and rs4979462) SNPs may confer susceptibility to systemic lupus erythematosus (SLE) risk, which were significantly associated with the clinical phenotypes of SLE. PMID: 29803925
  6. Three alternatively spliced isoforms of VEGI, VEGI174, VEGI192 and VEGI251 have been documented. This study investigated the effects of VEGI174 and its functional domains (V7 and V8) on epithelial-mesenchymal transition (EMT) in renal cell carcinoma (RCC) cells in vitro. Overexpression of VEGI174, V7 or V8 inhibited EMT. PMID: 28656288
  7. Results provide evidence that variance within TNFSF15 has the potential to affect cytokine expression across a range of tissues and thereby contribute to protection from infectious diseases such as leprosy, while increasing the risk of immune-mediated diseases including Crohn's disease and primary biliary cholangitis. PMID: 27507062
  8. single variant analysis detected a previously unreported psoriasis risk locus at TNFSF15 (rs6478108) PMID: 28973304
  9. play a role in the development of systemic sclerosis PMID: 28397078
  10. the DR3/TL1A pathway directly enhances human OC formation and resorptive activity, controlling expression and activation of CCL3 and MMP-9. PMID: 28062298
  11. the blocking of tumor necrosis factor receptor 2 (TNFR2) decreased TL1A-stimulated IL-6 production by rheumatoid arthritis fibroblast-like synoviocytes. PMID: 27081759
  12. Distinct but overlapping TNFSF15 haplotypes were demonstrated in diverticulitis patients versus healthy controls when compared with the known Crohn's risk haplotype suggesting similar but distinct genetic predispositions. This study strengthens the role for a genetic predisposition to diverticulitis that involves the TNFSF15 gene. PMID: 28624054
  13. TL1A differentially induces expression of TH17 effector cytokines IL-17, -9, and -22 and provides a potential target for therapeutic intervention in TH17-driven chronic inflammatory diseases. PMID: 27733581
  14. Our findings indicate that VEGI174 prevents progression and tumor metastasis through inhibiting epithelial-mesenchymal transition (EMT) in renal cell carcinoma (RCC) in vivo. This may provide a new approach for the treatment of RCC PMID: 28739718
  15. Data suggest that human regulatory T-lymphocytes express DR3 and demonstrate DR3/TL1A-mediated activation of signaling via MAP kinases and NFkappaB. (DR3 = death receptor 3; TL1A/TNFSF15 = tumor necrosis factor [ligand] superfamily, member 15) PMID: 28337757
  16. These results raise the possibility for involvement of TL1A/DR3/DR3-mediated mechanisms in epithelial-mesenchymal interactions and the development of inflammation-induced intestinal fibrosis in Crohn's disease. PMID: 27665176
  17. rs1250569 (ZMIZ1) and rs10114470 (TL1A) are two novel loci that indicate susceptibility to Inflammatory Bowel Disease in Han-Chinese patients. PMID: 28456797
  18. results support an idea that the genetic susceptibility of TNFSF15 to CD may be confounded, in part, by the increase of Prevotella PMID: 28197769
  19. (188)Re-NGR-VEGI has the potential as a theranostic agent. PMID: 26768609
  20. miRNA-31 can directly bind to the 3-UTR of TNFSF15, thereafter negatively regulating its expression in Caco2 cells. PMID: 27188743
  21. There were significant associations of rs3810936, rs6478108, rs6478109, rs7848647 with CD in Korean pediatric patients (P = 6.5x10(-8), P = 1.3x10(-8), P = 3.7x10(-8), P = 2.9x10(-8), respectively). PMID: 25998826
  22. Patients with mild traumatic brain injury (TBI) exhibited higher VEGI levels than those with moderate and severe TBI. PMID: 26945876
  23. Biologics beyond TNF-alpha inhibitors and the effect of targeting the homologues TL1A-DR3 pathway in chronic inflammatory disorders. PMID: 26810853
  24. Rs3810936 of TNFSF15 were related to the risk of ankylosing spondylitis PMID: 26823868
  25. Higher TL1A levels were associated with early stage chronic lymphocytic leukemia. PMID: 26393680
  26. TL1A-induced cell death is directly mediated through DR3. PMID: 26509650
  27. Plasma levels of TL1A were significantly higher in newly diagnosed SLE patients compared with controls, and were positively associated with SLE disease activity index. PMID: 25929716
  28. This study indicates that the HDAC inhibitor may be exploited as a therapeutic strategy modulating the soluble VEGI/DR3 pathway in osteosarcoma patients PMID: 25778932
  29. Results show that subjects with TNFSF15 -358CC genotype were at higher risks for developing gastric adenocarcinoma in the Helicobacter pylori infected group. PMID: 25251497
  30. The data indicate that TL1A may contribute to pathogenesis of inflammatory bowel diseases via local but not systemic induction of IL-17A but not IL-4, IL-13 or IFN-gamma. PMID: 26072972
  31. study has defined the increased serum and SF samples levels of TL1A and DcR3 in patients with rheumatoid arthritis (RA); findings support the hypothesis that TL1A and DcR3 may contribute to the pathogenesis of RA PMID: 25647275
  32. TNFSF15 SNPs, rs6478108 and rs4574921, may be independent genetic predictive factors for the development of stricture/non-perianal penetrating complications and perianal fistula, respectively. PMID: 24835165
  33. TL1A increases expression of CD25, LFA-1, CD134 and CD154, and induces IL-22 and GM-CSF production from effector CD4 T-cells PMID: 25148371
  34. Addition of TL1A to IL-1beta + IL-23 also augmented ILC3 proliferation PMID: 26046454
  35. This study shows an association between TNFSF15-rs3810936 and AAU and suggests that the TL1A/DR3 pathway may be implicated in the pathogenesis of this disease. PMID: 26200500
  36. associations exist between TNFSF15 gene polymorphisms and IBD (both CD and UC) in the Indian population PMID: 25501099
  37. These results suggested that TL1A could promote Th17 differentiation in rheumatoid arthritis via the activation of RORc, and this effect may be mediated by the binding of TL1A with DR3. PMID: 24832108
  38. TL1A blood levels are elevated in psoriasis patients; TL1A expression is higher in psoriatic lesions than in normal skin PMID: 25908025
  39. Human primary biliary cirrhosis-susceptible allele of rs4979462 enhances TNFSF15 expression by binding NF-1. PMID: 25899471
  40. Soluble TL1A synergized with IL-23 to stimulate peripheral blood mononuclear cells from patients with psoriasis vulgaris to produce IL-17. PMID: 25200589
  41. This meta-analysis indicated that most of the seven TNFSF15 polymorphisms (except for rs4263839) were risk factors contributed to CD and UC susceptibility. The differences in ethnicity did not influence the risk obviously. PMID: 25028192
  42. DR3 is expressed in some interstitial vascular endothelial cells (EC) in human kidney in situ; these EC also respond to its ligand TL1A by activating NF-kappaB. PMID: 25399326
  43. Mechanisms mediating TNFSF15:DR3 contributions to pattern recognition receptor outcomes included TACE-induced TNFSF15 cleavage to soluble TNFSF15; soluble TNFSF15 then led to TRADD/FADD/MALT-1- and caspase-8-mediated autocrine IL-1 secretion. PMID: 25197060
  44. This is the first report of the association between early Crohn's disease and the TNFSF15 single nucleotide polymorphisms. PMID: 25664710
  45. Tumor-infiltrating natural killer and CD4(+) T cells under the influence of cancer cells significantly increase the production of IFNgamma, which in turn inhibits TNFSF15 expression in vascular endothelial cells. PMID: 24141405
  46. may play an important role in the pathogenesis of primary biliary cirrhosis PMID: 24016146
  47. Our data demonstrate a key role for TL1A in promoting ILC2s at mucosal barriers. PMID: 24220298
  48. Combining the genetic marker TNFSF15 with ASCA IgA increased the power of predicting stenosis/perforating phenotype in Crohn's disease patients with TNFSF15 but not with a NOD2 genetic background PMID: 24783249
  49. genetic polymorphism is associated with psoriasis and psoriatic arthritis in Hungarians PMID: 24269700
  50. attenuated S. typhimurium carrying the dual function plasmid VEGI151/survivin cannot only be specifically enriched in the tumor tissue, but also showed a synergistic antitumor effect in vivo. PMID: 23404494

Show More

Hide All

Database Links

HGNC: 11931

OMIM: 604052

KEGG: hsa:9966

STRING: 9606.ENSP00000363157

UniGene: Hs.23349

Protein Families
Tumor necrosis factor family
Subcellular Location
Membrane; Single-pass type II membrane protein.; [Tumor necrosis factor ligand superfamily member 15, secreted form]: Secreted.
Tissue Specificity
Specifically expressed in endothelial cells. Detected in monocytes, placenta, lung, liver, kidney, skeletal muscle, pancreas, spleen, prostate, small intestine and colon.

Q&A

What is the molecular structure and function of human TNFSF15?

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 .

How is TNFSF15 expressed in human tissues and what regulates its expression?

TNFSF15 demonstrates a distinct expression pattern across human tissues:

  • Predominantly expressed in endothelial cells

  • Notably absent in B and T lymphocytes

  • Expression is inducible by TNF and IL-1α stimulation

  • 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.

What signaling pathways does TNFSF15 activate in different cell types?

Upon binding to its receptors, TNFSF15 activates several key signaling pathways with cell type-specific outcomes:

PathwayCellular EffectDownstream EffectsCell Types
NF-κBTranscription activationCytokine production, cell survivalMacrophages, T cells, endothelial cells
MAPKSignal transductionCell proliferation, gene expressionMultiple immune and non-immune cells
PI3KCell signalingCell growth, proliferationPrimarily immune cells
Caspase-8Proteolytic activationIL-1 secretion, pyroptosis regulationIntestinal myeloid cells, macrophages

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 .

What is the relationship between TNFSF15 and its receptors?

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 .

What are the optimal conditions for handling recombinant TNFSF15 in laboratory settings?

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.

What cell models are most appropriate for studying different TNFSF15 functions?

Selection of appropriate cell models depends on the specific TNFSF15 function under investigation:

Research FocusRecommended Cell ModelsRationale
Angiogenesis inhibitionHUVECs, HMEC-1Express TNFSF15 receptors; established models for angiogenesis studies
Immune modulationTHP-1, U937, primary macrophagesExpress DR3; suitable for studying cytokine responses
Inflammatory responsesPrimary intestinal myeloid cellsRelevant for IBD research; demonstrate TNFSF15-mediated cytokine secretion
Retinal protectionARPE-19, retinal endothelial cellsAppropriate for diabetic retinopathy studies

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

How can researchers effectively measure TNFSF15 activity in experimental systems?

Several complementary methodological approaches can be employed to assess TNFSF15 activity:

  • Signaling pathway activation:

    • Western blotting for phosphorylated NF-κB, MAPK, or PI3K components

    • Luciferase reporter assays for transcription factor activation

    • Phospho-flow cytometry for single-cell signaling analysis

  • 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:

    • Endothelial cell proliferation inhibition assays

    • Apoptosis detection methods (Annexin V/PI staining, TUNEL assay)

    • Pyroptosis assessment in relevant cell types (GSDME cleavage, LDH release)

  • Protein-protein interaction studies:

    • Co-immunoprecipitation of TNFSF15 with receptors or targets like GSDME

    • Surface plasmon resonance for binding kinetics analysis

    • Proximity ligation assays for visualizing interactions in situ

For comprehensive functional characterization, researchers should employ multiple complementary assays and include appropriate controls for each experimental approach.

What concentration ranges of recombinant TNFSF15 are effective for different experimental endpoints?

Optimal TNFSF15 concentrations vary based on the specific biological readout:

Experimental EndpointRecommended Concentration RangeTemporal Considerations
Cell signaling studies50-200 ng/mLAssess from 15 min to 24 h post-treatment
Cytokine induction100-500 ng/mLMeasure secretion 6-48 h after treatment
Angiogenesis inhibition200-1000 ng/mLEvaluate effects at 24-72 h
Cell death/survival10-500 ng/mLMonitor continuously over 24-72 h
Anti-pyroptotic effects100-500 ng/mLPre-treatment before pyroptotic stimulus

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.

How is TNFSF15 implicated in inflammatory bowel disease pathogenesis?

TNFSF15 has been identified as a significant risk factor in inflammatory bowel disease (IBD) through multiple mechanisms:

  • Genetic associations:

    • The TNFSF15 region is one of the 163 inflammatory bowel disease risk loci

    • Disease risk polymorphisms (e.g., rs6478108 A) are associated with increased TNFSF15 expression

    • Risk variants enhance pattern-recognition-receptor-induced signaling and cytokine secretion

  • Mechanistic contributions:

    • Enhanced TNFSF15:DR3 signaling amplifies PRR responses in macrophages

    • TACE-induced TNFSF15 cleavage leads to soluble TNFSF15 production

    • Soluble TNFSF15 promotes TRADD/FADD/MALT-1 and caspase-8-mediated IL-1 secretion

    • This creates a pro-inflammatory feedback loop in intestinal myeloid cells

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.

What role does TNFSF15 play in autoimmune disorders?

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:

    • TNFSF15-TNFRSF25 signaling promotes T-cell activation and proliferation

    • This pathway is essential for T-cell-mediated autoimmune responses

    • Through DR3 binding, TNFSF15 stimulates the generation of multiple cytokines

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.

How does TNFSF15 contribute to diabetic retinopathy protection?

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.

What is the relationship between TNFSF15 and asthma pathophysiology?

Emerging evidence suggests TNFSF15 may play a role in asthma development through its effects on angiogenesis:

  • Genetic associations:

    • Four SNPs in the TNFSF15 gene are associated with childhood-onset asthma in British white participants

    • The asthma-associated rs7856856 variant (T>C) is linked to reduced TNFSF15 gene expression

  • Proposed mechanism:

    • TNFSF15 functions as an inhibitor of vascular endothelial cell growth factor

    • Decreased TNFSF15 levels might contribute to increased angiogenesis in asthmatic patients

    • Angiogenesis is an important event in airway inflammation and tissue remodeling in asthma

  • Gene interaction network:

    • Genetic interactions are predicted between TNFSF15 with RORA, GATA3, and SLC8A1

    • Among 340 genes reported for asthma, 24 genes relate to cardiovascular system development

    • 14 of these genes associate with vasculature development, including 7 with angiogenesis

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.

How does TNFSF15 interact with pattern recognition receptors in inflammatory responses?

TNFSF15 plays a crucial role in amplifying pattern recognition receptor (PRR) responses through several mechanisms:

  • Enhancement of PRR signaling:

    • TNFSF15:DR3 interactions are critical for amplifying PRR-initiated MAPK/NF-κB/PI3K signaling

    • This leads to increased cytokine secretion in macrophages

    • Human macrophages clearly express DR3, enabling this response

  • Molecular mechanism:

    • The process involves TACE-mediated cleavage of TNFSF15 to produce soluble TNFSF15

    • Soluble TNFSF15 then triggers TRADD/FADD/MALT-1 and caspase-8-mediated pathways

    • This ultimately leads to autocrine IL-1 secretion

  • PRR types affected:

    • Broad range of pattern-recognition-receptors

    • Responses to mycobacterial components

    • Signaling induced by live bacteria

  • Disease relevance:

    • Enhanced in IBD risk variant carriers (rs6478108 A)

    • Contributes to intestinal inflammation

    • Represents a previously unknown mechanism through which TNFSF15:DR3 can contribute to inflammatory conditions

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.

What methodologies are effective for examining TNFSF15's role in angiogenesis?

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:

    • Models of diabetic retinopathy to study TNFSF15's protective effects

    • Laser-induced choroidal neovascularization

    • Tumor xenograft vascularization studies

  • 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 .

What techniques are effective for studying TNFSF15 polymorphisms and their functional effects?

Investigation of TNFSF15 polymorphisms requires comprehensive methodological approaches:

  • Genotyping strategies:

    • PCR-RFLP (polymerase chain reaction-restriction fragment length polymorphism)

    • Direct sequencing for verification

    • High-throughput genotyping platforms for population studies

  • Functional validation:

    • eQTL analysis to assess expression differences

    • Ex vivo stimulation assays with cells from genotyped individuals

    • Measurement of serum protein levels in different genotype carriers

  • 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.

How can the TNFSF15-DR3 signaling axis be targeted for therapeutic development?

The TNFSF15-DR3 signaling pathway presents several opportunities for therapeutic intervention:

  • Potential therapeutic strategies:

    • Recombinant TNFSF15 administration for conditions requiring angiogenesis inhibition or pyroptosis protection

    • TNFSF15-neutralizing antibodies for inflammatory conditions with elevated TNFSF15

    • Small molecule inhibitors of TNFSF15-DR3 interaction

    • Targeted modulation of downstream signaling components

  • Disease-specific approaches:

    • For diabetic retinopathy: Enhancing TNFSF15-GSDME interaction to prevent pyroptosis

    • For IBD: Inhibiting TNFSF15-induced amplification of PRR responses

    • For asthma: Potentially replacing deficient TNFSF15 to regulate angiogenesis

    • For SLE: Modulating TNFSF15 levels based on disease activity monitoring

  • 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.

How can contradictory findings about TNFSF15 function be reconciled in research?

Several apparent contradictions exist in TNFSF15 research literature that require careful interpretation:

  • Context-dependent effects:

    • TNFSF15 appears protective in diabetic retinopathy by inhibiting pyroptosis but pro-inflammatory in IBD by enhancing PRR signaling

    • Expression levels are increased in SLE but decreased in asthma

    • The same polymorphism may have different effects in different tissues

  • 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.

What considerations are important when designing experiments combining TNFSF15 with other immune modulators?

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:

    • TNFSF15 with TNF-α (both induce and respond to each other)

    • TNFSF15 with IL-1α (induces TNFSF15 expression)

    • TNFSF15 with pattern recognition receptor ligands (enhances responses)

    • TNFSF15 with VEGF (opposing effects on angiogenesis)

  • 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.

What are the best practices for maintaining recombinant TNFSF15 stability and activity?

Maintaining TNFSF15 stability is crucial for experimental reproducibility and reliable results:

  • Storage recommendations:

    • Store lyophilized protein at -20°C

    • For reconstituted protein, prepare single-use aliquots to avoid freeze-thaw cycles

    • Avoid repeated freeze/thaw cycles which compromise activity

  • Reconstitution best practices:

    • Use sterile PBS for initial reconstitution to 1 mg/mL

    • Allow complete dissolution before use

    • Consider adding carrier protein (0.1-1% BSA) for dilute solutions

    • Filter sterilize if needed using low protein-binding filters

  • 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.

How can researchers distinguish between membrane-bound and soluble TNFSF15 in experimental systems?

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:

    • Use of TACE/ADAM17 inhibitors to prevent cleavage of membrane-bound TNFSF15

    • Comparison of effects of cell-bound presentation versus soluble protein

    • Creation of cleavage-resistant TNFSF15 mutants

    • Use of specific blocking antibodies against different forms

  • Functional discrimination:

    • Soluble TNFSF15 leads to TRADD/FADD/MALT-1 and caspase-8-mediated IL-1 secretion

    • Membrane-bound forms may have distinct signaling properties

    • Different forms may preferentially activate different downstream pathways

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.

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.