The protein retains high biological activity, demonstrated through ligand-binding assays:
Crystallographic studies reveal TNFRSF9 forms a 2D network with TNFSF9, amplifying co-stimulatory signals in T cells . This structural insight supports its role in immune modulation.
TNFRSF9 is pivotal in cancer immunotherapy and autoimmune studies:
CAR-T Cell Therapy: Enhances T-cell persistence and anti-tumor efficacy by incorporating TNFRSF9 costimulation .
Bispecific Antibodies: Targets TNFRSF9 to activate tumor-infiltrating T cells while blocking PD-1/PD-L1 .
Structural Insights:
Clinical Relevance:
TNFRSF9, also known as 4-1BB, CD137, CDw137, and ILA (Induced By Lymphocyte Activation), is an inducible co-stimulatory receptor primarily expressed on activated T cells. It functions as a critical mediator in T cell activation, proliferation, differentiation, and apoptosis processes. TNFRSF9 also plays significant roles in the pathogenesis of certain autoimmune diseases and mediates anti-tumor immune responses of T cells and NK cells .
Researchers should be aware that literature searches may require using multiple nomenclatures to ensure comprehensive review of existing research. When reporting experimental findings, it is recommended to include all relevant nomenclatures in keywords and abstracts to enhance discoverability.
Commercially available recombinant human TNFRSF9 typically consists of amino acids Leu24-Gln186 of the native protein. The recombinant form often contains an N-terminal linker and a C-terminal tag (commonly a 10xHis-tag) when expressed in mammalian cells. The protein has an apparent molecular weight of approximately 28 kDa on gel electrophoresis and demonstrates high purity (>95%) with low endotoxin content (<1EU/μg) in research-grade preparations .
For functional validation, recombinant TNFRSF9 can bind to its ligand TNFSF9 with EC50 values ranging from 1.011-3.702 ng/mL in ELISA assays, confirming biological activity suitable for immune and antibody screening experiments .
Methodological approach for functional assessment:
Perform binding assays with natural ligand TNFSF9 using ELISA to determine EC50 values
Verify protein integrity via Western blot analysis under reducing and non-reducing conditions
Conduct cellular activation assays using TNFRSF9-responsive T cell lines
Analyze downstream signaling events (NF-κB activation, cytokine production)
Compare activity with established reference standards
Functional recombinant TNFRSF9 should demonstrate consistent binding kinetics with its ligand and induce expected biological responses in relevant cell systems .
TNFRSF9 deficiency presents a critical research model for understanding this receptor's immunological significance. Case studies of biallelic TNFRSF9 mutations reveal that CD137 deficiency severely compromises host defense against viral infections, particularly Epstein-Barr virus (EBV).
Immunological consequences include:
Markedly reduced or abrogated expression of CD137 on activated T, B, and NK cells
Impaired CD8+ T cell activation and function
Reduced expression and release of critical cytokines (IFN-γ, TNF-α)
Diminished expression of cytolytic molecules (perforin, granzyme B)
Compromised cytotoxic activity against infected cells
These observations highlight TNFRSF9's essential role in anti-viral immunity, particularly for controlling persistent viral infections. Researchers investigating primary immunodeficiencies should consider TNFRSF9 deficiency in patients presenting with severe EBV-associated lymphoproliferative disorders (LPD) .
To comprehensively investigate TNFRSF9 signaling pathways, researchers should employ a multi-modal approach:
Genetic manipulation techniques:
CRISPR/Cas9-mediated gene editing of TNFRSF9 or downstream effectors
Overexpression systems using wildtype or mutant TNFRSF9 constructs
siRNA/shRNA knockdown for transient signaling studies
Biochemical signaling analysis:
Phosphoproteomic analysis of downstream effectors (p38MAPK pathway)
Co-immunoprecipitation to identify novel interaction partners
Western blot analysis of activated signaling components
Functional readouts:
Cytokine production profiling (IFN-γ, TNF-α, IL-2)
Cytotoxicity assays against relevant targets
Proliferation and survival assays following TNFRSF9 engagement
Advanced imaging:
Live-cell imaging of TNFRSF9 clustering and immunological synapse formation
Super-resolution microscopy to visualize receptor distribution patterns
Research has established that TNFRSF9 signaling impacts p38MAPK phosphorylation, which subsequently regulates PAX6 expression. This signaling axis represents a targetable pathway in diseases where TNFRSF9 function is dysregulated .
TNFRSF9 demonstrates significant tumor-suppressive functions in several cancer models, particularly breast cancer. The mechanisms through which TNFRSF9 regulates tumor progression include:
Regulation of the p38MAPK/PAX6 signaling axis:
Expression patterns in tumor microenvironments:
Spatial expression patterns:
These findings position TNFRSF9 as a potential biomarker and therapeutic target in cancer research.
When investigating TNFRSF9 expression in tumor tissues, researchers should implement a comprehensive analysis strategy:
Tissue preparation and analysis:
Formalin-fixed paraffin-embedded (FFPE) and fresh-frozen tissue processing
Laser capture microdissection for isolation of specific cellular compartments
Multiplex immunohistochemistry to simultaneously assess TNFRSF9 expression with other markers
Spatial transcriptomics to map expression patterns relative to vascular structures and hypoxic regions
Expression quantification:
Digital pathology with automated quantification algorithms
Cell-type specific scoring systems (tumor cells vs. endothelial cells vs. immune infiltrates)
Gradient analysis to assess expression patterns relative to blood vessels or hypoxic regions
Correlation with clinical parameters:
Tumor size, grade, and stage
Mutation status of relevant cancer genes (e.g., BRAF V600E in melanoma)
Patient survival and treatment response data
Immune infiltration patterns and markers
Epigenetic analysis:
DNA methylation profiling of the TNFRSF9 promoter
Chromatin accessibility studies (ATAC-seq)
Histone modification analysis at the TNFRSF9 locus
This systematic approach enables comprehensive characterization of TNFRSF9 expression patterns in tumors and facilitates identification of clinically relevant associations .
TNFRSF9 (CD137/4-1BB) has emerged as a crucial component in second-generation chimeric antigen receptor (CAR) T cell therapy designs. Researchers should consider the following aspects when incorporating TNFRSF9 into CAR constructs:
Structural integration:
Position of the TNFRSF9 co-stimulatory domain relative to CD3ζ signaling domain
Optimization of linker sequences between domains
Potential for domain mutations to enhance or modulate signaling strength
Functional characteristics of TNFRSF9-containing CARs:
Enhanced T cell persistence compared to CD28-based CARs
Balanced effector-memory phenotype development
Reduced T cell exhaustion during repeated antigen exposure
Distinct cytokine production profiles compared to other co-stimulatory domains
Experimental validation approaches:
In vitro cytotoxicity against target cells expressing varying antigen densities
Long-term persistence assays in serial re-stimulation models
Detailed phenotypic characterization using multiparameter flow cytometry
In vivo efficacy and persistence in relevant preclinical models
Clinical translation considerations:
Optimized manufacturing protocols specific for TNFRSF9-containing CARs
Monitoring for unique toxicity profiles
Biomarkers to predict and assess response to TNFRSF9-based CAR-T cells
TNFRSF9 incorporation in CAR-T cells has demonstrated significant advantages for treating certain malignancies, particularly in terms of long-term persistence and memory formation, making it a valuable component in current CAR designs .
Several innovative approaches are being explored for therapeutic targeting of TNFRSF9 in cancer immunotherapy:
Agonistic antibodies:
Development of antibodies with optimal binding kinetics and epitope specificity
Engineering of antibody Fc regions to enhance or minimize FcγR engagement
Bispecific antibody formats linking TNFRSF9 engagement to tumor targeting
Targeted delivery strategies:
Antibody-cytokine fusion proteins (immunocytokines)
Tumor-targeting scFv:TNF fusion proteins that enhance TNFRSF9 signaling
Protease-activated prodrug approaches using tumor-selective proteases
Hypoxia-responsive TNFRSF9 targeting agents based on observed hypoxia-driven expression
Combination therapeutic approaches:
Synergistic pairing with checkpoint inhibitors (anti-PD-1, anti-CTLA-4)
Combination with conventional therapies (radiation, chemotherapy)
Sequential timing strategies to optimize immune activation
Coupling with NF-κB inhibitors or proteasome inhibitors to modulate signaling outcomes
Biomarker development:
TNFRSF9 DNA methylation as a predictive biomarker for immunotherapy response
Expression patterns on tumor and immune cells as response indicators
Soluble TNFRSF9 as a potential blood-based biomarker
These approaches highlight the complexity and potential of TNFRSF9-targeted immunotherapeutic strategies, with particular emphasis on overcoming previous clinical challenges related to toxicity and efficacy .
Genetic variations in TNFRSF9 have significant implications for protein function and disease susceptibility, as evidenced by recent discoveries:
Pathogenic mutations:
Biallelic heterozygous mutations (e.g., NM_001561.5: c.208+1→AT and c.452C>A [p.T151K]) can cause CD137 deficiency
These hypomorphic mutations result in markedly reduced or abrogated CD137 expression on activated immune cells
Functional consequences include impaired T cell activation, reduced cytokine production, and diminished cytotoxic activity
Clinical manifestations include increased susceptibility to severe EBV-associated lymphoproliferative disease
Experimental approaches to study genetic variations:
Site-directed mutagenesis to generate TNFRSF9 variants
Stable cell line creation expressing wild-type vs. mutant TNFRSF9
CRISPR/Cas9 knock-in models recapitulating human mutations
Patient-derived primary cell analyses
Structure-function relationship analyses:
Mapping mutations to functional domains
Computational modeling of protein structural changes
Ligand binding assays for variant proteins
Signaling pathway activation assessments
Clinical correlation strategies:
Genotype-phenotype correlation studies
Response to immunomodulatory therapies based on variant status
Development of personalized therapeutic approaches for patients with TNFRSF9 mutations
Understanding these genetic variations provides critical insights for researchers investigating primary immunodeficiencies and developing targeted therapeutics for patients with TNFRSF9-related disorders .
TNFRSF9 DNA methylation represents an emerging epigenetic biomarker with significant implications for cancer biology and immunotherapy response prediction:
Methylation analysis methodologies:
Bisulfite sequencing of TNFRSF9 promoter and regulatory regions
Methylation-specific PCR for targeted analysis
Genome-wide methylation arrays incorporating TNFRSF9 CpG sites
Integration with transcriptomic data to correlate methylation with expression
Cancer-specific methylation patterns:
Differential methylation between tumor and normal adjacent tissues
Correlation with TNFRSF9 expression levels in various tumor types
Association with tumor progression and clinical outcomes
Relationship to tumor immune microenvironment characteristics
Predictive value for immunotherapy:
Correlation between TNFRSF9 methylation status and response to immune checkpoint inhibitors
Integration into multiparameter predictive models
Potential as a companion diagnostic for TNFRSF9-targeted therapies
Longitudinal assessment during treatment to monitor response
Mechanistic insights:
Relationship between hypoxia and TNFRSF9 methylation
Impact of epigenetic modifying drugs on TNFRSF9 expression
Interaction with transcription factors and chromatin remodeling complexes
These findings provide rationale for further investigating TNFRSF9 DNA methylation as a predictive biomarker for immunotherapy response, potentially enabling more precise patient selection for various immunotherapeutic approaches .
Proper storage and handling of recombinant TNFRSF9 protein is critical for maintaining biological activity and experimental reproducibility:
Storage recommendations:
Reconstitution protocols:
Use sterile, filtered buffer to reconstitute lyophilized protein
Allow protein to equilibrate to room temperature before reconstitution
Gently mix by swirling rather than vortexing to prevent protein denaturation
Briefly centrifuge to collect all material at the bottom of the vial
Quality control measures:
Verify protein concentration after reconstitution
Perform activity testing using standardized binding assays
Check for aggregation using dynamic light scattering
Monitor endotoxin levels for experiments sensitive to bacterial contaminants
Shipping and transfer conditions:
Transport on blue ice to maintain temperature
Validate activity after shipping or transfer between laboratories
Document freeze-thaw cycles and storage durations
Adherence to these storage and handling guidelines will ensure optimal experimental outcomes when working with recombinant TNFRSF9 protein .
Investigation of the TNFRSF9/p38MAPK/PAX6 signaling axis requires a systematic experimental approach:
Pathway validation experiments:
siRNA/shRNA knockdown of TNFRSF9 followed by assessment of p-p38 and PAX6 levels
Overexpression of TNFRSF9 with concurrent monitoring of downstream components
P38 inhibitor studies (e.g., SB203580) to verify the dependence of PAX6 upregulation on p38 phosphorylation
Rescue experiments using constitutively active or dominant-negative constructs
Signaling kinetics analysis:
Time-course experiments following TNFRSF9 activation
Quantitative assessment of phosphorylation status at multiple timepoints
Single-cell analysis to account for heterogeneity in signaling responses
Correlation between signaling dynamics and functional outcomes
Cell models and conditions:
Comparison across multiple cell lines with varying baseline TNFRSF9 expression
Primary cells versus established cell lines
Consideration of microenvironmental factors (hypoxia, inflammatory cytokines)
3D culture systems to better recapitulate in vivo signaling
Functional readouts:
Cell proliferation, invasion, and apoptosis assays
Gene expression profiling to identify additional pathway components
Interaction with other signaling pathways (crosstalk mapping)
Systems biology approaches to model pathway dynamics
This comprehensive approach enables detailed characterization of this important signaling axis and provides opportunities to identify novel therapeutic targets for diseases where TNFRSF9 function is dysregulated .
Cutting-edge technologies are revolutionizing the study of TNFRSF9 function within complex tissue microenvironments:
Spatial multi-omics approaches:
Spatial transcriptomics to map TNFRSF9 expression patterns relative to vascular structures and cell types
Multiplexed ion beam imaging (MIBI) for simultaneous detection of multiple proteins
Imaging mass cytometry for high-dimensional spatial phenotyping
Digital spatial profiling to quantify protein and RNA abundance with spatial context
Advanced organoid and tissue models:
Patient-derived tumor organoids with preserved immune components
Microfluidic organ-on-chip systems modeling tumor-immune interactions
3D bioprinting of tissue models with controlled TNFRSF9 expression
Ex vivo tissue slice cultures maintaining native architecture
Single-cell analysis platforms:
Single-cell RNA sequencing with TNFRSF9 pathway gene signatures
CyTOF mass cytometry for high-parameter protein profiling
Cellular indexing of transcriptomes and epitopes (CITE-seq)
Spatial single-cell sequencing technologies
In vivo imaging technologies:
Intravital microscopy with fluorescently labeled TNFRSF9 antibodies
PET imaging with radiolabeled TNFRSF9-targeting agents
Photoacoustic imaging for deep tissue visualization
Bioluminescence resonance energy transfer (BRET) systems for pathway activation monitoring
These technologies provide unprecedented insights into TNFRSF9 biology within native tissue contexts, enabling more translational research approaches .
Optimization of TNFRSF9-based therapies requires innovative approaches to address current clinical limitations:
Reducing systemic toxicity:
Development of tumor-selective activation strategies
Incorporation of protease-sensitive linkers activated in tumor microenvironments
Engineering of prodrug-like antibody formats with inhibitory domains shed at tumor sites
Spatially restricted activation through tumor-targeting bispecific constructs
Enhancing efficacy:
Rational combination with checkpoint inhibitors or conventional therapies
Optimization of dosing schedules and administration routes
Development of TNFRSF9 agonists with optimized receptor crosslinking properties
Engineering of novel scaffold formats beyond traditional antibodies
Precision medicine approaches:
Implementation of TNFRSF9 methylation as a biomarker for patient selection
Development of companion diagnostics to identify optimal responders
Monitoring of tumor immune microenvironment changes during treatment
Adaptive trial designs with biomarker-guided treatment modifications
Manufacturing and stability improvements:
Enhanced production systems for complex biologics
Formulation optimization for improved stability and delivery
Development of extended-release or depot formulations
Novel delivery systems for improved tumor penetration