AITRL (Activation-Inducible TNF-Related Ligand) and GITRL (Glucocorticoid-Induced TNF-Related Ligand) refer to the same protein, designated as TNFSF18 in the TNF superfamily classification. This Type II single transmembrane protein shares relatively low sequence conservation with other TNFSF members. Human GITRL/AITRL cDNA encodes a 177 amino acid protein with the carboxy-terminal extracellular domain showing limited sequence identity to other family members: TNF/TNFSF2 (21%), Fas ligand/TNFSF6 (21%), TRAIL/TNFSF10 (18%), and lymphotoxin alpha/TNFSF1 (18%) .
Methodological approach: When investigating AITRL/GITRL, researchers should employ multiple detection techniques including ELISA, flow cytometry, and Western blotting to confirm identity and expression, as nomenclature variations can lead to confusion in literature searches and experimental design.
AITRL is primarily expressed on antigen-presenting cells including macrophages, immature and mature dendritic cells, and B cells. It is also constitutively expressed in human umbilical vein endothelial cells but is notably absent in resting or stimulated T cell lines, B cell lines, or peripheral blood mononuclear cells .
Methodological approach: For detecting cellular expression patterns, researchers should employ:
Flow cytometry with validated antibodies for surface expression
RT-PCR for transcriptional analysis
Immunohistochemistry for tissue localization
Single-cell RNA sequencing for heterogeneous populations
Unlike mouse GITR, human GITR expression is not induced by dexamethasone treatment but is upregulated following antigen-receptor stimulation or treatment with soluble anti-CD3 plus anti-CD28 or PMA plus ionomycin .
AITRL binds to its receptor AITR/GITR (TNFRSF18), which is expressed on T lymphocytes, natural killer (NK) cells, and certain antigen-presenting cells. The receptor is present at low levels in peripheral blood T cells, bone marrow, thymus, spleen, and lymph nodes . Upon binding by AITRL, AITR can be released from the cell surface. This receptor-ligand interaction induces nuclear factor (NF)-kappa B activation via TNF receptor-associated factor 2 and protects cells from TCR activation-induced cell death .
Methodological approach: Study of receptor-ligand interactions should include:
Co-immunoprecipitation assays
Surface plasmon resonance for binding kinetics
Reporter assays for downstream signaling activation
Proximity ligation assays for in situ detection
The structure-function relationship of AITRL is critical for its biological activity. Recent engineering approaches have developed HERA-GITRL, a hexavalent human GITR agonist designed as a single-polypeptide chain with three copies of GITRL receptor-binding domains fused to an IgG1-derived silenced Fc-domain that serves as a dimerization scaffold .
Methodological approach: Structural analysis should integrate:
X-ray crystallography or cryo-EM for 3D structure determination
Molecular modeling for interaction prediction
Mutagenesis studies to identify critical residues
Functional assays comparing different structural variants
The hexavalent structure of HERA-GITRL creates a spatially well-defined arrangement that clusters six receptor chains, inducing potent agonistic activity without requiring additional crosslinking. In direct in vitro comparison to a bivalent clinical benchmark anti-GITR antibody and a trivalent GITRL, only the hexavalent HERA-GITRL showed full biological activity independent of additional crosslinking .
T cell activation by AITRL involves complex signaling cascades that require systematic evaluation using complementary methods.
Methodological approach: A comprehensive experimental framework includes:
Activation Parameter | Recommended Assay | Timepoint |
---|---|---|
Surface markers | Flow cytometry (CD25, CD69) | 6-48 hours |
Cytokine production | ELISA/CBA (IL-2, IFN-γ) | 24-72 hours |
Proliferation | CFSE dilution or Ki67 staining | 3-5 days |
Signaling pathways | Phospho-flow (NF-κB, ERK) | 15-60 minutes |
Transcriptional changes | RNA-seq or qPCR arrays | 6-24 hours |
Metabolic reprogramming | Seahorse analysis (OCR/ECAR) | 24-48 hours |
Functional activity | Cytotoxicity or suppression assays | 4-72 hours |
When analyzing data, researchers should account for donor variability by using multiple donors and appropriate statistical methods for paired comparisons .
Scientific contradictions regarding AITRL function may arise from methodological differences, species-specific biology, or context-dependent effects.
Methodological approach: Researchers should adopt action research methodology with cyclical processes of planning, action, observation, and reflection . This approach includes:
Systematic documentation of experimental variables and conditions
Side-by-side comparisons using standardized protocols
Validation across multiple experimental systems
Integration of complementary techniques
Careful control of variables that might influence outcomes:
Cell activation status
Receptor density
Signal strength and duration
Microenvironmental factors
Genetic background
Experimental Variable | Control Strategy | Potential Impact |
---|---|---|
AITRL concentration | Dose-response curves | Threshold vs. graduated effects |
Timing of measurement | Time-course experiments | Transient vs. sustained responses |
Cell purity | Flow sorting > 95% purity | Elimination of bystander effects |
Receptor expression | Quantitative flow cytometry | Correlation of expression with function |
Genetic background | CRISPR controls | Validation of specificity |
Investigating AITRL in disease contexts requires thoughtful experimental design that balances clinical relevance with experimental rigor.
Methodological approach: For human disease studies, researchers should consider:
Ex vivo analysis of patient samples:
Paired comparison of affected vs. unaffected tissues
Longitudinal sampling during disease progression
Flow cytometric analysis of AITRL expression on immune cell subsets
Functional assays with patient-derived cells
Humanized mouse models:
NSG mice reconstituted with human immune components
PDX models with human tumor and immune cells
CRISPR-engineered models with human AITRL/AITR
In vitro disease modeling:
3D organoid cultures with immune components
Microfluidic systems for dynamic interactions
Co-culture systems with disease-relevant cell types
The action research process should include iterative cycles of experimental design, implementation, data analysis, and refinement of hypotheses .
Artificial intelligence offers powerful tools for extracting insights from complex AITRL experimental data.
Methodological approach: AI integration in AITRL research should follow this framework:
Data preparation:
Standardization and normalization
Feature extraction
Quality control metrics
Analysis approaches:
Supervised learning for predictive modeling
Unsupervised clustering for pattern discovery
Network analysis for pathway interactions
Natural language processing for literature mining
Validation strategies:
Cross-validation techniques
Independent dataset validation
Biological validation of computational predictions
AI applications can help identify optimal experimental conditions, predict AITRL interactions with other immune pathways, and design more efficient research protocols .
Translating AITRL research findings into clinical applications requires addressing multiple dimensions of drug development.
Methodological approach: Development strategy should include:
Target validation:
Genetic evidence (knockout/knockin models)
Expression correlation with disease outcomes
Functional validation in human samples
Therapeutic modality selection:
Efficacy assessment:
In vitro functional assays
Humanized mouse models
Ex vivo patient sample testing
Safety evaluation:
On-target/off-tumor effects
Cytokine release assessment
Autoimmunity risk analysis
Therapeutic Approach | Advantages | Methodological Considerations |
---|---|---|
Antibody-based | Clinical precedent, stability | Potential crosslinking requirements |
Recombinant proteins | Natural ligand mimicry | Shorter half-life, manufacturing complexity |
Hexavalent constructs | Enhanced receptor clustering | Novel format, immunogenicity risk |
Cell therapies | Targeted delivery, persistence | Complex manufacturing, heterogeneity |
Integrating multiple omics platforms provides comprehensive insights into AITRL biology in human contexts.
Methodological approach: Multi-omics integration should proceed as follows:
Coordinated sample collection:
Matched samples for different omics platforms
Preservation methods optimized for each analysis
Clinical annotation and metadata capture
Individual omics analyses:
Genomics: WGS, WES, or targeted sequencing
Transcriptomics: Bulk and single-cell RNA-seq
Proteomics: Mass spectrometry, proximity extension assays
Epigenomics: ATAC-seq, ChIP-seq, DNA methylation
Metabolomics: Targeted or untargeted metabolite profiling
Integration strategies:
Multi-omics factor analysis
Network-based data integration
Causal inference modeling
Trajectory analysis for dynamic processes
Validation approaches:
Orthogonal experimental validation
Independent cohort replication
Functional testing of predictions
Multi-omics analysis can reveal regulatory networks controlling AITRL expression, downstream effects of AITRL signaling, and potential biomarkers for therapeutic response .
Accurate detection and quantification of AITRL presents several technical challenges that require methodological solutions.
Methodological approach: Researchers should implement these strategies:
Antibody validation:
Knockout controls for specificity
Recombinant protein standards
Multiple antibody clones targeting different epitopes
Complementary detection methods:
Flow cytometry for cellular expression
ELISA for soluble forms
Mass cytometry for multi-parameter analysis
Imaging methods for tissue localization
Standardization approaches:
Quantitative flow cytometry with calibration beads
Absolute quantification using reference standards
Internal controls for inter-assay comparison
Emerging technologies:
Aptamer-based detection
Proximity-based assays
Single-molecule approaches
Researchers should document all validation steps and include appropriate controls in experimental design to ensure reproducibility and reliability of AITRL detection.
Species differences present significant challenges in AITRL research translation.
Methodological approach: To bridge animal and human studies, researchers should:
Develop comparative systems:
Side-by-side testing of human and animal AITRL
Cross-species reactive reagents when possible
Humanized animal models
Focus on conserved mechanisms:
Identify signaling pathways conserved across species
Target fundamental biological processes
Validate findings in human samples
Implement translational platforms:
Human ex vivo systems
Organoids with immune components
Microphysiological systems ("organs-on-chips")
Apply computational approaches:
Interspecies sequence and structure comparisons
Pathway conservation analysis
Predictive modeling of cross-species differences
The experimental approach should follow the cyclical pattern described in action research methodology, with continual refinement based on observational data .
The landscape of AITRL research is evolving with technological innovations that offer new experimental possibilities.
Methodological approach: Researchers should consider these emerging platforms:
CRISPR-based technologies:
Precise gene editing for mechanistic studies
CRISPRa/CRISPRi for expression modulation
CRISPR screens for pathway discovery
Base editing for specific mutations
Advanced imaging approaches:
Super-resolution microscopy for molecular interactions
Intravital imaging for in vivo dynamics
Multiplexed imaging for complex cellular interactions
Live-cell imaging for temporal processes
Single-cell technologies:
Multi-modal single-cell analysis (protein + RNA)
Spatial transcriptomics for tissue context
Single-cell functional assays
Lineage tracing for developmental processes
Bioinformatics and computational approaches:
Machine learning for experimental design
Network analysis for systems biology
Integrative multi-omics analysis
Virtual screening for therapeutic development
By applying these technologies within an action research framework, investigators can iteratively refine their understanding of AITRL biology and develop more effective interventions .
Understanding AITRL biology has significant implications for personalized therapeutic strategies.
Methodological approach: To advance precision medicine applications, researchers should:
Identify patient stratification biomarkers:
AITRL/AITR expression patterns
Genetic polymorphisms affecting the pathway
Functional assays predicting response
Combination biomarker panels
Develop companion diagnostics:
Flow cytometry-based assays
Tissue-based expression analysis
Soluble receptor/ligand quantification
Genetic testing for relevant variants
Design personalized therapeutic approaches:
Dose adjustment based on receptor density
Combinatorial strategies based on immune profiles
Timing optimization based on disease stage
Integration with other immunomodulatory agents
Implement adaptive trial designs:
Biomarker-guided patient selection
Early response assessment
Dynamic treatment allocation
Longitudinal monitoring protocols
This approach aligns with the action research methodology of planning, action, observation, and reflection in an iterative cycle to continuously improve therapeutic strategies .
Activation-Induced Tumor Necrosis Factor Receptor Ligand (AITRL), also known as Tumor Necrosis Factor Superfamily Member 18 (TNFSF18), is a crucial protein involved in immune modulation. AITRL is a member of the tumor necrosis factor superfamily and plays a significant role in regulating immune responses. The recombinant form of AITRL, produced in Escherichia coli, is widely used in research to study its biological functions and therapeutic potential.
AITRL is a transmembrane protein that consists of 177 amino acids, including a 28 amino acid cytoplasmic region, a 21 amino acid transmembrane domain, and a 128 amino acid extracellular domain . The recombinant human AITRL is typically produced in Escherichia coli as a single, non-glycosylated polypeptide chain containing 129 amino acids (72-199) with a molecular mass of approximately 14.6 kDa . The protein is purified using proprietary chromatographic techniques to achieve high purity levels, often greater than 90% as determined by SDS-PAGE .
AITRL is primarily expressed in endothelial cells and interacts with its receptor, Activation-Induced Tumor Necrosis Factor Receptor (AITR), also known as TNFRSF18 . This interaction leads to downstream signaling events that modulate immune cell function. AITRL plays a pivotal role in T-cell activation, proliferation, and differentiation. It can stimulate effector T-cell responses while also promoting the development and function of regulatory T cells, thus maintaining immune homeostasis .
The dysregulation of AITRL signaling has been implicated in various immune-related disorders, including autoimmune diseases, allergic reactions, and cancer . Due to its significant role in immune regulation, AITRL has emerged as a potential target for therapeutic interventions. Researchers are exploring the use of recombinant AITRL in developing treatments for these conditions, aiming to modulate immune responses effectively.