TNFSF18 binds to its receptor GITR (TNFRSF18), triggering immune-modulatory effects:
T-Cell Regulation: Enhances proliferation of CD4+/CD8+ T cells and natural killer (NK) cells while inhibiting regulatory T-cell (Treg) suppression .
Cytokine Production: Induces IFN-γ, IL-2, and IL-10 secretion in activated T cells .
Anti-Tumor Effects: Augments NK cell cytotoxicity and CD8+ T-cell responses against tumors .
Signaling Pathways: Activates NF-κB and STAT1, upregulating adhesion molecules (VCAM1, ICAM1) .
Functional assays demonstrate an EC₅₀ of 50.8 ng/mL in ELISA-based GITR binding and 2.56–2.94 ng/mL in TNFRSF18 interaction .
This protein is widely used in:
Immune Checkpoint Studies: Investigating GITR-TNFSF18 interactions for cancer immunotherapy .
Autoimmune Disease Models: Exploring its role in rheumatoid arthritis and ocular inflammation .
Endothelial Cell Activation: Studying leukocyte adhesion and monocyte migration .
Drug Development: Screening agonistic antibodies or small molecules targeting GITR .
Neutralizing soluble TNFSF18 enhances NK cell-mediated tumor lysis .
Agonistic GITR antibodies synergize with PD-1 inhibitors to suppress melanoma growth .
TNFSF18 overexpression in dendritic cells suppresses dexamethasone-induced immunosuppression .
In rheumatoid arthritis, TNFSF18 promotes macrophage adhesion and cytokine release (e.g., TNF-α, IL-6) .
GITRL (Glucocorticoid-induced TNF receptor ligand) is a cytokine that binds to TNFRSF18 (also known as AITR or GITR). It plays a crucial role in regulating T-cell responses, acting as a costimulator that lowers the activation threshold and enhances T-cell proliferation. GITRL is also important for interactions between activated T lymphocytes and endothelial cells. Its effects include activation of NF-κB, increased STAT1 phosphorylation, and upregulation of VCAM1 and ICAM1, promoting leukocyte adhesion to endothelial cells and regulating monocyte migration from the spleen to inflammatory sites.
GITRL's involvement in various diseases and biological processes is supported by extensive research:
TNFSF18, also known as Glucocorticoid-Induced TNF-Related Ligand (GITRL), is a type II transmembrane glycoprotein belonging to the tumor necrosis factor (TNF) superfamily. In scientific literature, it's also referred to as Activation-inducible TNF-related ligand (AITRL), TL6, and hGITRL in human contexts . The protein functions as a cytokine that binds to TNFRSF18/AITR/GITR and plays significant roles in regulating T-cell responses . The human TNFSF18 gene is located on chromosome 1q23 and encodes a protein with a molecular weight of approximately 22,724 Da .
Human TNFSF18/GITR Ligand has a characteristic structure consisting of:
A 50 amino acid cytoplasmic domain
A 21 amino acid transmembrane segment
Within the extracellular domain, human GITR Ligand shares 56% amino acid sequence identity with mouse GITR Ligand and 60% with rat GITR Ligand . Functionally, the protein forms homotrimers which is typical of the TNF superfamily proteins . The protein contains the characteristic TNF homology domain (THD) that is involved in both the formation of ligand trimers and in ligand-receptor interaction specificity .
TNFSF18 demonstrates a specific cellular expression pattern that is relevant to its immunological functions. It is predominantly expressed on:
Antigen-presenting cells (APCs)
B cells
Dendritic cells (DCs)
Macrophages
Endothelial cells
CD4-CD8- double negative thymic precursors
Neurons
Expression of TNFSF18 is not static but can be transiently upregulated in response to proinflammatory stimulation, suggesting its role in inflammatory responses . This dynamic expression pattern is important to consider when designing experiments involving inflammatory conditions or when using TNFSF18 as a marker for certain immune states.
TNFSF18 functions as the specific ligand for TNFRSF18, also known as Glucocorticoid-Induced TNF Receptor Family Related Protein (GITR) . GITR is constitutively expressed at high levels on regulatory T cells (Tregs) and at lower levels on resting CD25-CD4+ T cells, though expression increases markedly following T cell activation . The receptor is also found on other activated immune cells including NK cells and neutrophils .
The interaction between TNFSF18 and GITR leads to several immunological outcomes:
Costimulation of T cell activation and proliferation
Promotion of cytokine production
Enhanced expression of activation antigens
In murine models, abrogation of Treg-mediated suppression (though this effect appears species-specific and may not occur in humans)
Different expression systems yield recombinant TNFSF18 proteins with varying biological activities, which is critical knowledge for experimental design. Based on available data, there are notable differences:
The enhanced activity observed with certain recombinant forms suggests that oligomerization or specific conformational changes may influence receptor binding and downstream signaling. When selecting a recombinant TNFSF18 for research, investigators should consider whether their experimental questions require baseline or enhanced activity profiles .
The TNFSF18-GITR interaction activates multiple downstream signaling pathways that are crucial for its immunomodulatory functions:
NF-κB Pathway: TNFSF18 binding to GITR mediates activation of the NF-κB transcription factor . This can be measured through:
Nuclear translocation assays of NF-κB subunits
Luciferase reporter assays for NF-κB activity
Phosphorylation status of IκB proteins
STAT1 Phosphorylation: TNFSF18 triggers increased phosphorylation of STAT1 . Measurement approaches include:
Western blotting for phospho-STAT1
Flow cytometry using phospho-specific antibodies
Immunofluorescence microscopy to detect nuclear translocation
Adhesion Molecule Expression: TNFSF18 upregulates expression of VCAM1 and ICAM1 . This can be assessed by:
Flow cytometry of surface expression
qRT-PCR for transcript levels
Functional adhesion assays
When designing experiments to evaluate these pathways, time-course analyses are recommended as different pathways may activate with distinct kinetics following TNFSF18-GITR engagement.
Species-specific differences in TNFSF18 function present significant challenges for translational research:
Proper handling of recombinant TNFSF18 is critical for maintaining its biological activity. Based on manufacturer recommendations:
Reconstitution Protocol:
Reconstitute lyophilized TNFSF18 at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin .
Allow the protein to dissolve completely by gentle agitation.
Aliquot to minimize freeze-thaw cycles.
Storage Conditions:
Ship at ambient temperature
Upon receipt, store immediately at recommended temperature
Use a manual defrost freezer and avoid repeated freeze-thaw cycles
For long-term storage, keep at -20°C or -80°C depending on formulation
Stability Considerations:
Working stock solutions are typically stable for 1 month at 2-8°C
Carrier-free formulations may have different stability profiles than those containing carriers like BSA
Monitor protein activity periodically if stored for extended periods
Validating the biological activity of recombinant TNFSF18 is essential before using it in complex experimental systems. Several functional assays can be employed:
T Cell Proliferation Assay:
NF-κB Activation Assay:
Use reporter cell lines expressing GITR and an NF-κB responsive element driving luciferase expression
Treat with varying concentrations of TNFSF18
Measure luciferase activity after 6-24 hours
Expected result: Dose-dependent increase in reporter activity
Cytokine Production:
Stimulate primary T cells or NK cells with TNFSF18
Measure cytokine production (e.g., IFN-γ, IL-2) by ELISA or flow cytometry
Expected result: Enhanced cytokine production in TNFSF18-treated cultures
Treg Functional Assay (species-specific considerations required):
Detection of TNFSF18 requires different approaches depending on the experimental context:
Protein Detection Methods:
ELISA: Sandwich ELISA kits are commercially available with detection ranges of 78-5000 pg/mL and sensitivity around 20 pg/mL
Western Blotting: Use reducing conditions; molecular weight approximately 22.7 kDa
Flow Cytometry: Surface staining for membrane-bound form
Immunohistochemistry/Immunofluorescence: For tissue localization studies
mRNA Detection:
qRT-PCR: Design primers specific to human TNFSF18 (avoid regions with high homology to other TNF family members)
RNA-Seq: For transcriptome-wide analysis of TNFSF18 expression in different cell populations
In situ hybridization: For spatial expression analysis in tissues
Special Considerations:
When detecting recombinant tagged proteins, antibodies against the tag (e.g., His, HA) can be used
For detecting both membrane-bound and soluble forms, choose antibodies recognizing the extracellular domain
When working with primary human samples, account for potential polymorphisms that might affect antibody binding
Contradictory findings regarding TNFSF18 function are common in the literature and can arise from several factors:
Species-Specific Differences:
The most notable contradiction involves Treg suppression, which is abrogated by TNFSF18-GITR interaction in mice but not in humans
Methodological approach: Always clearly specify the species origin of both the TNFSF18 protein and target cells in publications
Interpretation strategy: Consider evolutionary differences in immune regulation between species when comparing results
Context-Dependent Signaling:
TNFSF18 can induce different outcomes depending on:
Cell activation state (resting vs. activated)
Cell type (T cells vs. APCs)
Presence of other costimulatory signals
Methodological approach: Include appropriate controls for cell activation state and document all experimental conditions thoroughly
Membrane-Bound vs. Soluble Forms:
Membrane-bound TNFSF18 may signal differently than recombinant soluble forms
Interpretation strategy: Consider whether observed effects might differ between natural membrane-bound TNFSF18 and recombinant soluble forms
Standardized Analysis Framework:
When comparing contradictory results, create a table documenting:
Species/cell origin
TNFSF18 form used (membrane vs. soluble, tagged vs. untagged)
Readout systems
Experimental conditions
This systematic approach helps identify variables that might explain seemingly contradictory outcomes
Robust experimental design for TNFSF18 research requires specific controls:
Protein-Specific Controls:
Heat-inactivated TNFSF18 (to control for non-specific protein effects)
Isotype-matched control protein (for tagged recombinant proteins)
Blocking antibodies against TNFSF18 or GITR (to confirm specificity of observed effects)
Cell-Specific Controls:
GITR-deficient cells (to confirm receptor dependency)
Comparison of effects on different T cell subsets (CD4+ vs. CD8+, naive vs. memory)
Inclusion of relevant APCs when studying T cell responses
Activation Controls:
Submaximal TCR stimulation conditions (where TNFSF18 costimulatory effects are most evident)
Positive controls using established costimulatory molecules (e.g., anti-CD28)
Time-course experiments to capture both early and late events
Signaling Pathway Controls:
Specific pathway inhibitors (e.g., NF-κB inhibitors) to confirm involvement of hypothesized mechanisms
Phosphorylation state analysis of multiple pathway components
Recombinant TNFSF18 offers multiple approaches to investigate and manipulate Treg function, though with important species-specific considerations:
Mouse Models (where TNFSF18-GITR interaction abrogates Treg suppression):
In vitro applications:
Addition of recombinant TNFSF18 to Treg suppression assays (expected outcome: reduced suppression)
Pretreatment of isolated Tregs before functional assays
Dose titration to establish threshold effects
In vivo applications:
Administration of recombinant TNFSF18 or agonistic anti-GITR antibodies
Development of TNFSF18-expressing cell-based therapies
TNFSF18-Fc fusion proteins with extended half-life
Human Systems (where direct abrogation of Treg suppression is not observed):
Focus on TNFSF18's costimulatory effects on effector T cells
Investigate whether indirect mechanisms might influence Treg/Teff balance
Examine effects on Treg stability and phenotype rather than suppressive function
Methodological Recommendations:
Always include parallel mouse and human experiments when studying Treg effects
Thoroughly characterize Treg phenotype (FOXP3, CD25, GITR expression) before and after TNFSF18 treatment
Assess multiple suppression mechanisms (cytokine production, metabolic inhibition, direct contact)
TNFSF18 has significant implications for inflammation and autoimmunity research due to its immunoregulatory properties:
Endothelial Cell Interactions:
TNFSF18 is important for interactions between activated T lymphocytes and endothelial cells
It triggers increased phosphorylation of STAT1 and upregulates expression of VCAM1 and ICAM1
These effects promote leukocyte adhesion to endothelial cells and regulate migration of monocytes from the splenic reservoir to sites of inflammation
Research application: Study TNFSF18 as a potential target for modulating vascular inflammation
Reverse Signaling in APCs:
Autoimmune Disease Models:
Dysregulation of TNFSF18 has been implicated in various autoimmune diseases
Research applications include:
Studying TNFSF18 expression patterns in autoimmune disease tissues
Correlating TNFSF18 levels with disease severity or treatment response
Developing targeted therapies that modulate the TNFSF18-GITR axis
Several significant limitations exist in current TNFSF18 research:
Species-Specific Differences:
The functional divergence between human and mouse TNFSF18-GITR systems complicates translational research
Solution approach: Development of humanized mouse models specifically for studying the human TNFSF18-GITR axis
Structural Insights:
Limited high-resolution structural data on TNFSF18-GITR complexes hampers structure-based drug design
Solution approach: Prioritize crystallography or cryo-EM studies of the complex to guide development of more specific modulators
Physiological Relevance:
Most studies use recombinant soluble TNFSF18, which may not fully recapitulate the biology of membrane-bound forms
Solution approach: Develop systems that present TNFSF18 in membrane-bound format (e.g., cell-based assays, liposome-bound protein)
Context-Dependent Function:
TNFSF18 effects are highly dependent on the broader immunological context
Solution approach: More comprehensive systems biology approaches that examine TNFSF18 function in concert with other immunoregulatory pathways