TNFSF9 binds to TNFRSF9 (4-1BB/CD137), a costimulatory receptor expressed on activated T cells, NK cells, and antigen-presenting cells (APCs) . Key functions include:
T Cell Activation: Promotes proliferation, survival, and cytokine production (e.g., IL-2, IFN-γ) in CD4⁺/CD8⁺ T cells, enhancing antitumor immunity .
Reverse Signaling: On APCs, induces inflammatory cytokines (e.g., TNF-α, IL-6) and modulates macrophage polarization .
Oncogenic Roles: In pancreatic cancer, TNFSF9 activates Wnt/β-catenin/Snail pathways, driving epithelial-mesenchymal transition (EMT) and metastasis .
Assay Type | EC₅₀ Value | Application | Source |
---|---|---|---|
Functional ELISA | 2.67–3.70 ng/mL | TNFRSF9 binding validation | |
T Cell Proliferation | Not reported | Co-stimulatory signal induction |
Therapeutic Potential:
Cancer Immunotherapy: Agonistic antibodies targeting TNFRSF9 are in clinical trials for solid tumors, leveraging TNFSF9-TNFRSF9 signaling to enhance T cell responses .
Autoimmune Diseases: Dual role observed; may exacerbate CD8⁺ T cell-mediated inflammation or suppress CD4⁺ T cell-driven autoimmunity .
Pancreatic Cancer: TNFSF9 overexpression correlates with poor prognosis. It promotes metastasis via:
Contradictory Roles: In hepatocellular carcinoma and colorectal cancer, TNFSF9 inhibits proliferation, highlighting context-dependent effects .
TNFSF9-knockdown pancreatic cancer cells show reduced M2 markers (IL-10, TGF-β) and increased M1 markers (TNF-α, IL-8) in co-culture experiments with macrophages .
TNFSF9, also known as 4-1BB Ligand or CD137L, is a type II transmembrane protein belonging to the tumor necrosis factor (TNF) superfamily that plays a crucial role in immune response activation. The protein consists of an intracellular domain, a transmembrane segment, and an extracellular domain (ECD) that mediates its biological activity. TNFSF9 binds to its receptor TNFRSF9 (4-1BB/CD137) which is expressed on activated CD4+ and CD8+ T cells, thymocytes, NK cells, monocytes, neutrophils, dendritic cells, and eosinophils . Upon binding, TNFSF9 transduces a co-stimulatory signal that promotes proliferation, activation, and survival of CD4+ and CD8+ T cells .
The protein plays a distinct temporal role in T cell responses, with CD28 being important for initial T cell expansion, while TNFSF9-TNFRSF9 interactions act later in the response . Additionally, this signaling supports the survival and responsiveness of memory T cells during viral infection . TNFSF9 can also induce reverse signaling in monocytes to stimulate inflammatory cytokine production, and in macrophages, TNFSF9 associates with TLR4 to enhance inflammatory responses .
TNFSF9 exists in both membrane-bound and soluble forms, each with distinct biological properties that researchers must consider when designing experiments. The membrane-bound form is approximately 50 kDa and consists of an 82 amino acid cytoplasmic domain, a 21 amino acid transmembrane segment, and a 206 amino acid extracellular domain in rat models . A smaller, 26 kDa soluble form can be released from the surface of activated cells while retaining bioactivity .
For experimental applications, recombinant forms typically consist of the extracellular domain with various tags. For example, commercially available human TNFSF9 may include the region from amino acids 71-254 with an N-terminal tag . When designing experiments, researchers should carefully consider which form best represents their biological question of interest. Membrane-bound TNFSF9 studies may require cell-based systems, while soluble TNFSF9 effects can be studied using recombinant proteins in solution.
The optimal reconstitution protocol for recombinant TNFSF9 depends on the specific formulation. For lyophilized preparations, the following methodology is recommended:
Briefly centrifuge the vial prior to opening to bring contents to the bottom
Reconstitute in deionized sterile water
For carrier-containing formulations, reconstitute at 250 μg/mL in PBS
For carrier-free formulations, reconstitute in PBS with consideration for the final application
Consider adding glycerol (5-50% final concentration) if preparing for long-term storage
For optimal results, avoid repeated freeze-thaw cycles by preparing single-use aliquots. When working with reconstituted protein, maintain at 4°C for up to one week . For experimental consistency, verify protein concentration after reconstitution using appropriate protein assay methods.
TNFSF9 expression has significant associations with tumor immune microenvironment (TIME) characteristics, particularly in renal cell carcinoma (RCC). High TNFSF9 expression correlates with several important immune parameters:
Gene pathway analysis reveals that tumors with high TNFSF9 expression show upregulation of adaptive immune responses, response to bacteria, and extracellular matrix-associated proteins . This suggests that TNFSF9-high tumors feature enhanced antigen presentation and regulated immune responses. Interestingly, despite having increased CD8+ T cells and interferon-gamma levels, these tumors also contain more dysfunctional T cells and regulatory T cells (Tregs) . This seemingly contradictory finding suggests a complex immune regulatory environment where both immune activation and suppression mechanisms are enhanced.
The study of TNFSF9's role in T-B cell interactions requires specialized methodologies that can detect subtle changes in cellular interactions and activation states. Based on research findings, the following approaches are recommended:
Immune Cell Population Analysis: CIBERSORT algorithm analysis reveals that TNFSF9-high tumors have increased T follicular helper (TFH) cells and plasma B cells, suggesting enhanced T-B interactions . Researchers should employ multiparameter flow cytometry with markers for TFH cells (CXCR5, PD-1, ICOS) and B cell subsets.
Functional Co-stimulation Assays: To measure TNFSF9's co-stimulatory effects, researchers can use recombinant TNFSF9 in combination with anti-CD3 stimulation to assess IL-2 secretion by T cells. The ED50 for this effect is typically 5-30 ng/mL . This approach isolates TNFSF9-specific effects from other co-stimulatory pathways.
Binding Interaction Studies: Characterize the binding interactions between TNFSF9 and its receptor using functional ELISA methods. Typical protocols involve coating TNFRSF9 at 100 ng/mL and measuring binding of recombinant TNFSF9 with an ED50 of 1-6 ng/mL .
Gene Expression Pathway Analysis: Tools like Metascape can identify upregulated adaptive immune response pathways in TNFSF9-high samples . This approach contextualizes TNFSF9's role within broader immune signaling networks.
TNFSF9 shows significant potential as a predictive biomarker for immunotherapy response, particularly for patients receiving combination nivolumab plus ipilimumab (NIVO+IPI) therapy for metastatic renal cell carcinoma (mRCC). The methodology for developing and utilizing TNFSF9 as a biomarker includes:
Expression Analysis: In clinical studies, TNFSF9 expression discriminated between response and non-response groups with 88.89% sensitivity and 87.50% specificity (AUC = 0.9444) . This outperformed PD-L1 expression (AUC = 0.75) .
Threshold Determination: Establishing expression thresholds that optimize predictive value requires ROC curve analysis with validation across multiple cohorts.
Multiparameter Integration: Combining TNFSF9 with other potential biomarkers (TAP1, CD8A) may enhance predictive power .
Implementation Protocol:
Collect tumor tissue samples prior to immunotherapy initiation
Measure TNFSF9 expression using validated RNA-seq or immunohistochemistry methods
Classify patients as high or low TNFSF9 expressors based on established thresholds
Integrate with clinical decision-making for therapy selection
This approach leverages the finding that TNFSF9-high tumors demonstrate enhanced adaptive immune responses, increased T and B cell infiltration, and particular sensitivity to CTLA-4 inhibition due to their elevated Treg populations .
Carrier proteins significantly impact the stability and experimental utility of recombinant TNFSF9. Bovine Serum Albumin (BSA) is commonly added as a carrier protein to enhance stability, increase shelf-life, and allow storage at more dilute concentrations . The effects of carrier proteins include:
Parameter | With Carrier (BSA) | Carrier-Free | Recommendation |
---|---|---|---|
Stability | Enhanced | Reduced | Use carrier-containing formulations for general applications |
Shelf-life | Extended | Shorter | Use carrier-containing formulations for long-term studies |
Storage Concentration | Can be more dilute | Requires higher concentration | Consider experimental needs for protein concentration |
Experimental Interference | Potential interference in some assays | Minimal interference | Use carrier-free for applications where BSA could interfere |
Formulation | Lyophilized from PBS with BSA | Lyophilized from PBS with Trehalose | Select based on downstream application |
For cell or tissue culture applications and ELISA standards, the BSA-containing formulation is generally recommended . In contrast, carrier-free protein is preferable for applications where BSA might interfere with the experimental system . Researchers should carefully consider these factors when selecting recombinant TNFSF9 preparations for their specific experimental designs.
Detecting soluble TNFSF9 (sTNFSF9) in experimental and clinical samples requires specialized methodologies due to its relatively low abundance (26 kDa form) compared to the membrane-bound version . The following approaches have proven effective:
ELISA-Based Detection:
Sandwich ELISA using capture and detection antibodies specific to TNFSF9
Sensitivity can be enhanced using biotin-streptavidin amplification systems
Typical detection ranges are in the pg/mL to ng/mL range
Western Blot Analysis:
Sample concentration may be required prior to electrophoresis
Use reducing conditions to distinguish monomeric sTNFSF9
Molecular weight markers should focus on the 25-30 kDa range
Functional Binding Assays:
Measure binding to recombinant TNFRSF9 proteins coated on plates
Assess biological activity through downstream signaling events
Clinical Sample Processing:
For serum/plasma: collect in appropriate anticoagulant tubes and process within 30 minutes
Centrifuge at 1000-2000g for 10 minutes at 4°C
Store aliquots at -80°C to prevent degradation
Avoid repeated freeze-thaw cycles
When interpreting results, researchers should consider that a 26 kDa soluble form of TNFSF9 can be released from activated cells while retaining bioactivity . This soluble form may have distinct biological effects compared to the membrane-bound version.
TNFSF9 expression significantly impacts patient response to combination immunotherapy, particularly the nivolumab plus ipilimumab (NIVO+IPI) regimen in metastatic renal cell carcinoma. Research findings demonstrate several key relationships:
Predictive Value: TNFSF9 expression discriminates between responders and non-responders with 88.89% sensitivity and 87.50% specificity (AUC = 0.9444), outperforming PD-L1 as a biomarker (AUC = 0.75) .
Immune Microenvironment Characteristics: High TNFSF9 expression correlates with:
T Cell Function: TNFSF9-high tumors show complex T cell dynamics:
Therapeutic Mechanism Insight: The increased Treg population in TNFSF9-high tumors suggests why dual checkpoint inhibition (NIVO+IPI) is particularly effective. While PD-1 inhibition (nivolumab) addresses T cell exhaustion, CTLA-4 inhibition (ipilimumab) likely suppresses Treg activation, creating a synergistic effect specifically beneficial in TNFSF9-high tumors .
These findings suggest that TNFSF9 expression analysis could be integrated into clinical decision-making to identify patients most likely to benefit from combination immunotherapy approaches.
Modeling TNFSF9-dependent immune activation in cancer immunotherapy research requires specialized experimental systems that recapitulate the complex interactions between tumor cells, antigen-presenting cells, and effector T cells. Based on the current understanding of TNFSF9 biology, the following experimental systems are recommended:
3D Co-culture Systems:
Composition: Tumor cells, dendritic cells, T cells
Rationale: Models TNFSF9-mediated interactions between antigen-presenting cells and T cells in a tumor-like context
Readouts: T cell proliferation, cytokine production, tumor cell killing
Humanized Mouse Models:
Implementation: Immune-deficient mice reconstituted with human immune cells
Advantages: Allows study of human TNFSF9-TNFRSF9 interactions in vivo
Assessment Methods: Flow cytometry of tumor-infiltrating lymphocytes, cytokine profiling, tumor growth kinetics
Ex Vivo Tumor Slice Cultures:
Approach: Fresh tumor tissue maintained in culture with preserved architecture
Benefits: Maintains intact tumor microenvironment with endogenous TNFSF9 expression
Analyses: Immunohistochemistry, flow cytometry, cytokine profiling
In Vitro Functional Assays:
When designing experiments, researchers should consider that TNFSF9's effects may differ depending on the phase of immune response, as it acts later than CD28 co-stimulation in T cell responses . Additionally, both membrane-bound and soluble forms of TNFSF9 (26 kDa) retain bioactivity and may have distinct effects .
The interaction between TNFSF9 and its receptor TNFRSF9 involves specific molecular binding events that trigger downstream signaling cascades critical for immune cell activation. Key aspects of this interaction include:
Binding Kinetics and Affinity:
Structural Requirements:
The extracellular domain of TNFSF9 contains the receptor-binding region
In recombinant systems, the region from amino acids 71-254 (for human TNFSF9) maintains biological activity
Proper folding is essential for activity, with proper disulfide bond formation crucial for maintaining the active conformation
Receptor Expression and Distribution:
TNFRSF9 (4-1BB/CD137) is expressed on activated CD4+, CD8+, memory CD8+, NKT, and regulatory T cells
It is also found on myeloid and mast cell progenitors, dendritic cells, mast cells, and bacterially infected osteoblasts
This distribution pattern defines the cell types responsive to TNFSF9 signaling
Functional Consequences of Binding:
TNFSF9-TNFRSF9 binding transduces a co-stimulatory signal that promotes T cell proliferation, activation, and survival
It supports the survival and responsiveness of memory T cells during viral infection
Unlike CD28 co-stimulation which is important for initial T cell expansion, TNFSF9-TNFRSF9 signaling acts later in the immune response
Understanding these molecular interactions is crucial for designing experiments to study TNFSF9 function and for developing therapeutic approaches targeting this pathway.
TNFSF9 activates distinct signaling pathways in different immune cell populations, leading to context-dependent functional outcomes. These pathway activations have significant implications for experimental design and interpretation:
T Cells:
Monocytes and Macrophages:
Dendritic Cells:
Expression Patterns: Found on activated dendritic cells
Role: Influences maturation and antigen presentation capabilities
Myeloid Progenitors:
These diverse signaling events highlight the need for cell-type-specific experimental approaches when studying TNFSF9 function. Researchers should carefully consider which cell populations and readouts are most relevant to their specific research questions about TNFSF9 biology.
Working with recombinant TNFSF9 presents several technical challenges that researchers should anticipate and address through careful experimental design:
Protein Stability Issues:
Carrier Protein Interference:
Challenge: BSA carrier in standard preparations may interfere with certain assays
Solution: Use carrier-free preparations (typically stabilized with trehalose instead of BSA) for applications where carrier might interfere
Applications: Carrier-free is preferred for binding assays, certain cell culture systems, and in vivo studies
Reconstitution Difficulties:
Activity Variations Between Preparations:
Challenge: Functional activity may vary between lots or preparations
Solution: Validate each new lot with functional assays such as binding to TNFRSF9 or T cell co-stimulation assays
Expected Values: ED50 for binding to TNFRSF9 should be 1-6 ng/mL; for T cell co-stimulation (IL-2 secretion) should be 5-30 ng/mL
Endotoxin Contamination:
By addressing these common challenges, researchers can maximize the reliability and reproducibility of experiments involving recombinant TNFSF9.
Differentiating between the effects of membrane-bound and soluble TNFSF9 requires specialized experimental approaches that can isolate and compare these distinct forms:
Expression System Selection:
Experimental Separation Strategies:
Physical Separation: Use transwell systems to separate TNFSF9-expressing cells from target cells while allowing soluble factors to diffuse
Molecular Engineering: Create mutant forms that cannot be cleaved from the membrane or that are constitutively secreted
Selective Inhibition: Use metalloprotease inhibitors to prevent cleavage of membrane-bound TNFSF9
Functional Comparison Methodologies:
Direct Comparison: Set up parallel experiments with recombinant soluble TNFSF9 versus fixed TNFSF9-expressing cells
Dose-Response Analysis: Compare concentration-dependent effects of soluble TNFSF9 with density-dependent effects of membrane-bound TNFSF9
Temporal Dynamics: Assess differences in signaling kinetics and durability between the two forms
Analytical Considerations:
Western Blotting: Detect distinct molecular weight forms (approximately 50 kDa for membrane-bound versus 26 kDa for soluble)
Flow Cytometry: Quantify surface-expressed versus secreted TNFSF9 using specific antibodies
ELISA: Measure soluble TNFSF9 concentration in culture supernatants or biological fluids
Understanding the differential effects of these forms is critical as the 26 kDa soluble form released from activated cells retains bioactivity but may have distinct functional properties compared to the membrane-bound form .