TNFRSF4 is a co-stimulatory receptor in the tumor necrosis factor receptor superfamily. Its extracellular domain contains four cysteine-rich repeats, enabling binding to its ligand, OX40L (TNFSF4), expressed on activated antigen-presenting cells (APCs) and endothelial cells . Upon engagement, TNFRSF4 recruits TRAF2, TRAF3, and TRAF5, activating downstream signaling pathways like NF-κB and PI3K, which promote T-cell proliferation, cytokine production, and survival .
Key Functional Roles:
T-cell memory formation: Enhances transition from effector to memory T cells .
Th1/Th2 responses: Supports both arms of adaptive immunity .
Regulatory T cells (Tregs): Modulates Treg function and immune tolerance .
TNFRSF4 has emerged as a therapeutic target in autoimmune diseases and cancer.
Autoimmune Diseases:
Agonistic antibodies (e.g., OX86) enhance T-cell activation, while blocking antibodies (e.g., GSK3174998) suppress inflammation, showing efficacy in animal models of asthma and graft-versus-host disease .
Cancer Immunotherapy:
Tumors exploit TNFRSF4+ Tregs to evade immune detection. Targeting TNFRSF4 with antibodies reduces Treg function, boosting anti-tumor immunity .
Dual blockade of TNFRSF4 and PD-1/PD-L1 pathways synergistically improves anti-tumor responses .
Antibodies targeting TNFRSF4 are used in research and clinical trials for their ability to modulate immune responses.
Key Features:
Ber-ACT35: Recognizes an extracellular epitope of TNFRSF4, used to detect activated T cells in autoimmune diseases .
AF3388: Detects TNFRSF4 in human Tregs and PBMCs, validated in PHA-treated cells .
FAB1256P: Mouse-specific antibody for studying T-cell activation in murine models .
Recent studies highlight TNFRSF4's role in immune regulation and disease:
Leukemia:
TNFRSF4+ Tregs promote immune escape of leukemia stem cells (LSCs). Antibody-mediated inhibition of TNFRSF4 signaling enhances anti-leukemic immunity .
Cancer Immunotherapy:
Agonistic antibodies (e.g., OX86) combined with CpG oligonucleotides activate tumor-infiltrating T cells, improving tumor control in preclinical models .
Autoimmune Diseases:
TNFRSF4 (also known as OX40 or CD134) is a type I transmembrane glycoprotein of approximately 50 kDa that is expressed primarily on activated T cells . It functions as a co-stimulatory molecule within the tumor necrosis factor receptor superfamily.
The significance of TNFRSF4 stems from its critical role in:
T cell activation and proliferation
Generation of memory T cells
Regulation of T cell-mediated immune responses
Potential applications as an immunotherapeutic target
Recent research has highlighted TNFRSF4 as a promising biomarker in various cancers, particularly in relation to tumor microenvironment (TME) assessment and prognosis prediction .
TNFRSF4 antibodies serve multiple research applications, each requiring specific considerations:
The selection should be guided by the specific research question, sample type, and anticipated downstream analyses.
Validation of TNFRSF4 antibody specificity is crucial for experimental reliability. A comprehensive validation approach should include:
Positive and negative controls:
Transfected cell line testing:
Antibody isotype controls:
Cross-reactivity assessment:
Test against intended species (Human, Non-Human Primate) and verify specificity
Consider epitope conservation when transitioning between model systems
Multiple detection methods:
Confirm target detection using at least two independent techniques (e.g., flow cytometry and Western blot)
Proper validation ensures confidence in observed results and minimizes experimental artifacts.
TNFRSF4 antibodies have emerged as valuable tools for characterizing tumor microenvironment (TME) components, particularly immune infiltrates. Methodological approaches include:
Multiplex immunohistochemistry (m-IHC):
Flow cytometric analysis of tumor-infiltrating lymphocytes (TILs):
Utilize PE, FITC, or APC-conjugated TNFRSF4 antibodies in multi-parameter panels
Include markers for T cell subsets (CD3, CD4, CD8), activation status, and exhaustion
This approach allows quantification of TNFRSF4+ immune cells within the TME
Correlation with clinical outcomes:
Deconvolution algorithms:
These methods collectively provide insights into how TNFRSF4 contributes to immune surveillance and anti-tumor responses.
Flow cytometric analysis using TNFRSF4 antibodies requires careful attention to several key methodological aspects:
Sample preparation optimization:
Antibody selection and panel design:
Protocol optimization:
Titrate antibodies to determine optimal concentration
Include appropriate compensation controls
Use viability dyes to exclude dead cells, which can bind antibodies non-specifically
Gating strategy recommendations:
Data interpretation considerations:
Compare expression levels between activated and non-activated T cells
Assess co-expression with other activation markers
Consider both percentage of positive cells and mean fluorescence intensity
Following these guidelines ensures robust and reproducible flow cytometry results when studying TNFRSF4 expression patterns.
Different TNFRSF4 antibody clones recognize distinct epitopes and demonstrate varying functional characteristics that affect their research applications:
Key considerations for clone selection:
Epitope accessibility:
Certain epitopes may be masked in fixed tissues or denatured samples
Conformation-dependent epitopes may be lost in Western blotting
Functional requirements:
Cross-reactivity needs:
When working with non-human primates, Ber-ACT35 provides consistent results
For studies requiring broader species coverage, OX40-3108 offers advantages
Understanding these differences enables researchers to select the most appropriate clone for specific experimental requirements.
TNFRSF4 has emerged as a potential prognostic biomarker in several cancers, particularly endometrial cancer. To effectively evaluate its expression in clinical specimens:
Tissue microarray (TMA) analysis approaches:
Use standardized IHC protocols with validated antibodies
Implement quantitative scoring systems based on staining intensity and percentage of positive cells
Consider automated image analysis for objective quantification
Correlation with clinicopathological features:
Prognostic assessment methodology:
Apply disease-specific survival (DSS) analysis, defined as time between surgery and cancer-related death
Utilize Kaplan-Meier analysis with log-rank tests to evaluate prognostic impact
Consider multivariate Cox regression to adjust for confounding factors
Integration with immune marker profiles:
Evaluate TNFRSF4 in conjunction with CD4, CD8, and FOXP3
Calculate correlation coefficients (Spearman or Pearson) between TNFRSF4 and immune markers
This approach provides insights into the immunological context of TNFRSF4 expression
Meta-analysis of expression data:
These methodological approaches enable robust evaluation of TNFRSF4 as a prognostic biomarker in clinical research contexts.
Neutralization assays are crucial for evaluating the functional activity of TNFRSF4-targeting therapeutic approaches. A methodological framework includes:
Cell line selection and preparation:
Use HT1080 human fibrosarcoma cells transfected with human TNFRSF4
Maintain stable expression through appropriate selection markers
Include wild-type HT1080 cells as negative controls
Stimulation protocol:
Neutralization assay setup:
Pre-incubate cells with increasing concentrations of anti-TNFRSF4 antibody
Add recombinant OX40L/TNFSF4 at the optimized concentration
Include appropriate controls:
Isotype control antibody
Unstimulated cells
Stimulated cells without neutralizing antibody
Readout measurements:
Quantify CXCL8/IL-8 using ELISA (e.g., Human CXCL8/IL-8 Quantikine ELISA Kit)
Calculate neutralization dose (ND50) - the antibody concentration that inhibits 50% of cytokine production
For reference, clone 977974 has an ND50 of 0.6-3.0 μg/mL , while polyclonal antibodies may show ND50 of 0.025-0.16 μg/mL
Data analysis:
Generate dose-response curves using non-linear regression
Compare neutralization efficacy between different antibody clones
Assess correlation between epitope binding and neutralization capacity
This framework allows systematic evaluation of antibody-mediated TNFRSF4 neutralization, which is particularly relevant for therapeutic development.
Researchers frequently encounter specific technical challenges when working with TNFRSF4 antibodies. Here are methodological solutions:
Low signal intensity in flow cytometry:
High background in immunohistochemistry:
Multiple bands in Western blot:
Cross-reactivity in multi-species studies:
Inconsistent neutralization results:
Cause: Variability in recombinant protein activity or cell responsiveness
Solution: Establish dose-response curves for each new lot of recombinant OX40L/TNFSF4
Control: Include a standard neutralizing antibody with known ND50 as reference
These troubleshooting approaches enhance experimental reliability when working with TNFRSF4 antibodies across different applications.
Multiplex immunohistochemistry (m-IHC) with TNFRSF4 antibodies requires careful optimization for successful simultaneous detection of multiple markers. Key methodological considerations include:
Panel design strategy:
Sample preparation optimization:
For FFPE tissues: Antigen retrieval conditions critically affect TNFRSF4 detection
Test different retrieval methods (citrate pH 6.0 vs. EDTA pH 9.0)
Optimize fixation protocols to preserve epitope integrity
Sequential staining approach:
Order antibodies from lowest to highest abundance target
Consider placing TNFRSF4 early in the sequence if expression levels are low
Include thorough washing steps between antibodies to prevent cross-reactivity
Signal amplification methods:
For low-abundance TNFRSF4: Implement tyramide signal amplification (TSA)
Carefully titrate primary antibodies to determine optimal concentration
Include single-color controls to assess bleed-through and optimize exposure settings
Validation and controls:
Recommended antibody combinations:
TNFRSF4 (clone Ber-ACT35) + CD4 + CD8 + FOXP3 for T cell subset analysis
TNFRSF4 + proliferation markers (Ki67) + activation markers for functional assessment
This optimization framework enables robust m-IHC protocols that can reveal the spatial and functional relationships between TNFRSF4+ cells and other immune components within tissues.