TNFRSF25 (also known as DR3, LARD, or TRAMP) is a transmembrane receptor with a death domain, primarily expressed in lymphoid cells. The biotin-conjugated TNFRSF25 antibody is a polyclonal or monoclonal antibody chemically linked to biotin, facilitating its use in techniques such as Western blot (WB), immunohistochemistry (IHC), and flow cytometry .
Gene ID: 8718
Synonyms: DR3, LARD, TRAMP, Apo-3, TNFRSF12
Function: Regulates lymphocyte homeostasis, apoptosis, and NF-κB signaling .
Role in Asthma: TNFRSF25/TL1A signaling drives IL-13 production by NKT cells and Th2-polarized CD4+ T cells, exacerbating allergic lung inflammation. Blocking TNFRSF25 with antibodies reduces eosinophil infiltration and mucus production in murine asthma models .
Inflammatory Bowel Disease: TNFRSF25 expression correlates with disease severity, suggesting its utility as a biomarker .
Co-Stimulation of T Cells: Agonistic TNFRSF25 antibodies enhance CD8+ T cell activation and antitumor responses. Their efficacy depends on Fcγ receptor interactions (e.g., FcγRIIB/III) .
Multimeric TL1A Fusion Proteins: Engineered TL1A (TNFRSF25 ligand) proteins co-stimulate T cells independently of Fcγ receptors, offering a novel therapeutic avenue .
Mouse DR3: Detected with ~5% cross-reactivity to human DR3 in WB .
Biotin Compatibility: Compatible with streptavidin-HRP or fluorophore systems for signal amplification .
Therapeutic Potential: While biotin-conjugated TNFRSF25 antibodies are primarily diagnostic tools, agonistic anti-TNFRSF25 antibodies (non-biotinylated) show promise in cancer immunotherapy by enhancing T cell co-stimulation .
Safety Profile: TNFRSF25-targeted therapies exhibit minimal toxicity in preclinical models, supporting further development .
TNFRSF25, also known as DR3 (Death Receptor 3), is a T-cell co-stimulatory receptor belonging to the tumor necrosis factor receptor superfamily. In humans, the canonical protein consists of 417 amino acid residues with a molecular mass of 45.4 kDa . TNFRSF25 is primarily expressed in thymocytes and lymphocytes, making it a significant marker for immunological studies . Its importance stems from its role as a costimulatory receptor that influences both CD4+ and CD8+ T cell responses to cognate antigens . TNFRSF25 has garnered increased research interest due to its potential applications in cancer immunotherapy, as agonists targeting this receptor have demonstrated promising antitumor effects in preclinical studies .
Biotin conjugation of TNFRSF25 antibodies offers several methodological advantages:
Signal amplification: The strong affinity between biotin and streptavidin (Ka ≈ 10^15 M^-1) enables researchers to enhance detection sensitivity through secondary labeling with streptavidin-conjugated fluorophores or enzymes.
Experimental flexibility: Biotin-conjugated antibodies can be integrated into various detection systems including flow cytometry, immunohistochemistry, ELISA, and Western blotting, providing versatility across experimental platforms .
Multiplexing capability: When combined with differently conjugated antibodies, biotin-TNFRSF25 antibodies facilitate multi-parameter analysis in complex immunological studies.
Compatibility with avidin-biotin complex (ABC) methods: This enables signal enhancement in immunohistochemical applications, particularly valuable when studying tissues with low TNFRSF25 expression levels.
The strategic advantage of biotin conjugation becomes particularly evident when examining samples with low receptor expression or when multiple detection parameters are required simultaneously.
For optimal flow cytometry results with biotin-conjugated TNFRSF25 antibodies, follow this methodological approach:
Sample preparation:
Staining protocol:
Block non-specific binding with 5% normal serum in PBS for 30 minutes at 4°C
Add biotin-conjugated anti-TNFRSF25 antibody at optimized concentration (typically 0.1-1 μg per 10^6 cells)
Incubate for 30-45 minutes at 4°C in the dark
Wash twice with FACS buffer (PBS + 2% FBS + 0.1% sodium azide)
Add fluorophore-conjugated streptavidin (dilution typically 1:500-1:2000)
Incubate for 30 minutes at 4°C in the dark
Wash twice with FACS buffer
Acquire data on flow cytometer
Multiparameter considerations:
The biotin-streptavidin system offers signal amplification compared to direct fluorophore conjugation, which is particularly advantageous when studying regulatory T cells where TNFRSF25 may be expressed at variable levels.
For Western blot optimization with biotin-conjugated TNFRSF25 antibodies, implement the following protocol:
Sample preparation:
Extract proteins from cells/tissues using RIPA buffer containing protease inhibitors
Quantify protein concentration using Bradford or BCA assay
Prepare samples with reducing buffer containing DTT or β-mercaptoethanol
Electrophoresis considerations:
Transfer and blocking:
Transfer proteins to PVDF membrane (superior to nitrocellulose for glycosylated proteins)
Block with 5% BSA in TBST (preferred over milk for phosphoprotein detection)
Antibody incubation:
Dilute biotin-conjugated anti-TNFRSF25 antibody (typically 1:500-1:2000)
Incubate overnight at 4°C
Wash 3×10 minutes with TBST
Incubate with HRP-conjugated streptavidin (typically 1:5000-1:10000) for 1 hour at room temperature
Wash 3×10 minutes with TBST
Detection specificity considerations:
To validate results, consider testing crude cell lysates alongside membrane-enriched fractions, as TNFRSF25 is a transmembrane protein.
To enhance specificity in immunohistochemistry with biotin-conjugated TNFRSF25 antibodies:
Sample preparation protocol:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin
Section at 4-6 μm thickness
Perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Blocking endogenous biotin:
This critical step prevents false positives
Apply avidin solution for 15 minutes
Rinse with PBS
Apply biotin solution for 15 minutes
Rinse with PBS
Immunostaining procedure:
Block endogenous peroxidase with 3% H₂O₂ for 10 minutes
Apply protein block (2.5% normal horse serum) for 20 minutes
Incubate with biotin-conjugated anti-TNFRSF25 antibody (1:100-1:500) overnight at 4°C
Wash with PBS
Apply HRP-conjugated streptavidin for 30 minutes
Develop with DAB substrate
Counterstain with hematoxylin
Controls for validation:
Include thymus or lymphoid tissues as positive controls
Use isotype-matched biotin-conjugated control antibodies
Implement tissue-specific absorption controls to confirm specificity
Interpretation guidelines:
This approach enhances specificity by addressing the common challenges of endogenous biotin and cross-reactivity in immunohistochemical applications.
Biotin-conjugated TNFRSF25 antibodies provide valuable tools for investigating T cell costimulation in vaccine research through these methodological approaches:
Ex vivo T cell costimulation assays:
Isolate CD4+ and CD8+ T cells from vaccinated subjects
Immobilize biotin-conjugated TNFRSF25 antibodies on streptavidin-coated plates
Measure proliferation via [³H]-thymidine incorporation or CFSE dilution
Quantify cytokine production using multiplex bead arrays
Assess activation markers (CD25, CD69) via flow cytometry
Combined costimulation strategies:
Adjuvant development protocol:
Regulatory T cell modulation:
Biotin-conjugated antibodies enable precise tracking of Treg responses
Monitor the balance between effector and regulatory responses
Quantify the differential expansion of Tregs versus conventional T cells
The following table summarizes the comparative effects of TNFRSF25 and TNFRSF4 costimulation in vaccine contexts:
| Parameter | TNFRSF25 Costimulation | TNFRSF4 Costimulation |
|---|---|---|
| CD8+ T cell proliferation | Strong enhancement | Strong enhancement |
| CD4+ Tconv proliferation | Weak costimulation | Potent costimulation |
| Treg proliferation | Strong costimulation | Weak costimulation |
| IgG1 production | Enhanced | Minimal effect |
| IgG2a production | Enhanced | Moderately enhanced |
| IgG2b production | Enhanced | Minimal effect |
| Memory response | Affects secondary immunity | Affects secondary immunity |
This methodological approach provides insights into how TNFRSF25 targeting can enhance vaccine efficacy through costimulatory mechanisms .
When employing biotin-conjugated TNFRSF25 antibodies in cancer immunotherapy research, consider these advanced methodological approaches:
Tumor microenvironment analysis protocol:
Obtain tumor samples and prepare single-cell suspensions
Stain with biotin-conjugated TNFRSF25 antibodies alongside tumor-infiltrating lymphocyte markers
Analyze TNFRSF25 expression on CD8+ T cells, CD4+ conventional T cells, and regulatory T cells
Recent research indicates that while TNFRSF25 agonists expand splenic Treg cells, they may not significantly affect intratumoral Treg cells
Agonistic antibody development considerations:
Combination therapy experimental design:
Test biotin-conjugated TNFRSF25 antibodies in combination with:
Checkpoint inhibitors (anti-PD-1, anti-CTLA-4)
Chemotherapy agents
Radiation therapy
Monitor synergistic effects on tumor regression and immune memory formation
Analysis of long-term antitumor immunity:
Comparative analysis with TL1A-based approaches:
These methodological considerations address the complexities of TNFRSF25 targeting in cancer immunotherapy and provide a framework for robust experimental design.
For investigating TNFRSF25 isoform-specific functions using biotin-conjugated antibodies, implement this advanced methodological framework:
Isoform identification protocol:
Epitope mapping methodology:
Determine specific epitopes recognized by various biotin-conjugated TNFRSF25 antibodies
Create a panel of antibodies targeting different domains:
Extracellular domain (ECD)
Transmembrane domain
Intracellular death domain
Map epitope accessibility in different cellular contexts
Isoform-specific detection strategy:
Develop Western blot protocols optimized for resolving closely-sized isoforms:
Use gradient gels (4-15%) for improved separation
Employ longer running times at lower voltage
Consider 2D electrophoresis (IEF followed by SDS-PAGE)
Validate isoform identity using mass spectrometry
Functional analysis methodology:
Perform selective knockdown of specific isoforms using siRNA
Re-express individual isoforms in knockout models
Compare costimulatory capacity of different isoforms using T cell activation assays
Assess differential effects on:
Conventional T cells vs. regulatory T cells
CD4+ vs. CD8+ T cell responses
Cytokine production profiles
Post-translational modification analysis:
This comprehensive approach enables researchers to dissect the specific contributions of individual TNFRSF25 isoforms to receptor function, advancing our understanding of the molecular mechanisms underlying TNFRSF25 biology.
When working with biotin-conjugated TNFRSF25 antibodies in flow cytometry, researchers may encounter several technical challenges that can be methodically addressed:
Low signal intensity:
Problem: TNFRSF25 expression may be low in resting lymphocytes
Solution: Stimulate cells with PMA and calcium ionophore for 24 hours to upregulate TNFRSF25 expression
Methodology: Incubate cells in complete media with 50 ng/mL PMA and 1 μg/mL ionomycin
Expected outcome: 2-5 fold increase in staining intensity
High background staining:
Problem: Endogenous biotin in certain cell types
Solution: Implement biotin blocking steps
Protocol:
Incubate cells with unconjugated streptavidin (0.5 mg/mL) for 15 minutes
Wash cells twice with staining buffer
Add biotin (1 mg/mL) for 15 minutes to saturate remaining streptavidin binding sites
Wash cells twice before adding biotin-conjugated TNFRSF25 antibody
Nonspecific binding:
Problem: Fc receptor-mediated binding
Solution: Use appropriate blocking reagents
Implementation: Incubate cells with Fc block (anti-CD16/CD32) at 1 μg per 10^6 cells for 10 minutes prior to antibody staining
Signal interference in multiparameter analysis:
Problem: Spectral overlap between fluorophores
Solution: Optimize fluorophore selection for streptavidin conjugates
Strategy:
Choose streptavidin conjugated to fluorophores in spectral regions distinct from other markers
Perform proper compensation using single-stained controls
Consider using quantum dots conjugated to streptavidin for reduced spectral overlap
Variable detection in different lymphocyte subsets:
Problem: Heterogeneous expression levels
Solution: Subset-specific analysis approach
Implementation:
Gate on specific cell populations (CD4+, CD8+, Tregs)
Adjust PMT voltages to accommodate expression range
Compare median fluorescence intensity rather than percent positive
The following table summarizes troubleshooting approaches for different cell types:
| Cell Type | Common Issue | Optimization Strategy | Expected Improvement |
|---|---|---|---|
| Resting T cells | Low TNFRSF25 expression | PMA/ionomycin stimulation (24h) | 2-5× signal increase |
| Regulatory T cells | Variable expression levels | CD25/FoxP3 co-staining | Improved population resolution |
| Memory T cells | Background autofluorescence | Use streptavidin-APC or PE | Better signal-to-noise ratio |
| Activated T cells | Receptor internalization | Fixed/permeabilized staining | Detection of total protein |
| Thymocytes | High endogenous biotin | Extensive biotin blocking | Reduced background |
These methodological approaches ensure robust and reliable detection of TNFRSF25 across various experimental contexts .
To effectively validate the functional activity of biotin-conjugated TNFRSF25 antibodies in agonist versus antagonist studies, implement this comprehensive validation methodology:
Receptor binding validation:
Flow cytometry competitive binding assay:
Pre-incubate cells with unlabeled antibody
Add biotin-conjugated antibody
Measure displacement to confirm shared epitope binding
Surface plasmon resonance (SPR) analysis:
Measure binding kinetics (kon, koff, KD)
Compare with reference antibodies or natural ligand (TL1A)
Agonistic activity assessment protocol:
In vitro T cell costimulation assay:
Co-stimulate T cells with anti-CD3 and biotin-conjugated TNFRSF25 antibody
Measure proliferation (CFSE dilution or [³H]-thymidine incorporation)
Quantify activation markers (CD25, CD69)
Assess cytokine production (IL-2, IFN-γ)
NF-κB reporter assay:
Transfect cells with TNFRSF25 and NF-κB reporter construct
Stimulate with biotin-conjugated TNFRSF25 antibody
Measure luciferase activity
Compare with positive control (TL1A)
Antagonistic activity assessment:
Ligand blocking assay:
Pre-incubate cells with biotin-conjugated TNFRSF25 antibody
Add TL1A ligand
Measure inhibition of TL1A-induced effects
Functional inhibition assay:
Assess ability to block T cell costimulation
Measure reduction in cytokine responses
Quantify inhibition of NF-κB activation
Cross-validation with alternative formats:
Compare biotin-conjugated antibody with:
Unconjugated antibody
Alternative conjugates (fluorophore-conjugated)
F(ab')2 fragments (to eliminate Fc effects)
This approach distinguishes between conjugation-dependent and intrinsic antibody properties
In vivo functional validation:
The following data table illustrates typical validation parameters:
| Validation Parameter | Agonistic Profile | Antagonistic Profile | Neutral Binding |
|---|---|---|---|
| T cell proliferation | >200% of control | <80% of TL1A response | 80-120% of control |
| NF-κB activation | >2.5-fold increase | <0.5-fold of TL1A | No significant change |
| Treg expansion | Significant increase | No effect or decrease | No significant change |
| Cytokine production | IL-2, IFN-γ increase | Inhibition of TL1A-induced | No significant change |
| Antitumor activity | Tumor regression | No effect or promotion | Variable effects |
This comprehensive validation approach ensures accurate classification of biotin-conjugated TNFRSF25 antibodies as agonists or antagonists, critical for their application in immunotherapy research .
Several advanced methodologies are emerging to enhance the utility of biotin-conjugated TNFRSF25 antibodies in single-cell analysis:
Mass cytometry (CyTOF) integration:
Methodology: Combine biotin-conjugated TNFRSF25 antibodies with metal-tagged streptavidin
Advantages:
Eliminates spectral overlap issues of fluorescence
Enables simultaneous analysis of >40 parameters
Reduces autofluorescence background
Application: Comprehensive immune profiling of TNFRSF25+ cells within heterogeneous populations
Single-cell RNA sequencing coupled with protein detection:
Methodology: CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing)
Tag biotin-conjugated TNFRSF25 antibodies with oligonucleotide barcodes
Capture both transcriptome and surface protein expression simultaneously
Research value: Correlate TNFRSF25 protein levels with gene expression patterns at single-cell resolution
Application: Identify transcriptional signatures associated with TNFRSF25-expressing cell subsets
Spatial transcriptomics with antibody detection:
Methodology:
Apply biotin-conjugated TNFRSF25 antibodies to tissue sections
Detect with streptavidin-conjugated fluorophores
Overlay with spatial transcriptomics data
Research advantage: Preserve spatial context of TNFRSF25-expressing cells within the tissue microenvironment
Application: Mapping TNFRSF25+ cell distribution in lymphoid organs and tumor microenvironments
High-throughput imaging cytometry:
Methodology: Imaging Flow Cytometry
Combine biotin-conjugated TNFRSF25 antibodies with fluorescent streptavidin
Capture morphological features alongside marker expression
Research value: Assess TNFRSF25 cellular localization and internalization kinetics
Application: Track receptor dynamics following ligand engagement or antibody-mediated clustering
Proximity ligation assays for protein-protein interactions:
Methodology:
Combine biotin-conjugated TNFRSF25 antibodies with antibodies against potential interaction partners
Apply DNA-conjugated streptavidin and secondary antibodies
Ligate and amplify DNA when proteins are in close proximity
Research advantage: Detect and quantify TNFRSF25 interactions with signaling molecules in situ
Application: Map TNFRSF25 signaling networks in different T cell subsets
These emerging methodologies significantly expand the research applications of biotin-conjugated TNFRSF25 antibodies beyond conventional approaches, enabling more sophisticated investigations of TNFRSF25 biology at the single-cell level.
Biotin-conjugated TNFRSF25 antibodies offer several methodological approaches for developing innovative combination immunotherapies:
Dual-targeted immunotherapeutic approaches:
Methodology: Co-targeting TNFRSF25 with checkpoint inhibitors
Combine biotin-conjugated TNFRSF25 agonistic antibodies with anti-PD-1/PD-L1
Monitor synergistic activation of tumor-infiltrating lymphocytes
Research rationale: TNFRSF25 costimulation may counteract T cell exhaustion
Expected outcome: Enhanced CD8+ T cell effector function despite checkpoint inhibition
Bioorthogonal click chemistry applications:
Methodology:
Modify biotin-conjugated TNFRSF25 antibodies with azide groups
Separately administer drugs conjugated to cyclooctyne groups
Allow in vivo click reaction for targeted drug delivery
Research advantage: Precise delivery of payloads to TNFRSF25-expressing cells
Potential applications:
Targeted delivery of TLR agonists
Localized cytokine delivery
Selective delivery of small molecule immunomodulators
Bifunctional antibody development:
Methodology:
Create bifunctional molecules with biotin-conjugated TNFRSF25 binding domain
Combine with domains targeting tumor-associated antigens
Research advantage: Brings TNFRSF25+ T cells into proximity with tumor cells
Predicted outcome: Enhanced tumor-specific T cell activation
TNFRSF25-targeted nanoparticle delivery systems:
Methodology:
Functionalize nanoparticles with streptavidin
Attach biotin-conjugated TNFRSF25 antibodies
Load nanoparticles with immunomodulatory compounds
Research value: Controlled release of immunomodulators to TNFRSF25+ cells
Applications:
Delivery of siRNA targeting immunosuppressive pathways
Controlled release of costimulatory cytokines
Targeted delivery of epigenetic modifiers
Combination with cancer vaccines:
The following table summarizes potential combination strategies and their predicted outcomes:
These innovative approaches position biotin-conjugated TNFRSF25 antibodies as valuable tools in the development of next-generation combination immunotherapies with potentially superior efficacy profiles .
When selecting biotin-conjugated TNFRSF25 antibodies for research applications, investigators should prioritize these critical methodological considerations:
Epitope specificity and functional relevance:
Select antibodies targeting functionally relevant epitopes based on research goals:
Agonistic activity requires specific epitopes, as demonstrated by research showing that antibody 1A6-m1 exhibits greater antitumor activity than higher affinity antibodies targeting overlapping epitopes
For detection purposes, antibodies recognizing conserved epitopes across species may be preferable
Validate epitope specificity through competitive binding assays
Cross-reactivity with orthologs:
Biotin conjugation chemistry and density:
Assess the biotinylation method used:
NHS-ester chemistry (targets lysine residues)
Sulfhydryl-reactive maleimide chemistry (targets reduced cysteines)
Consider biotin-to-protein ratio:
Optimal range typically 3-6 biotin molecules per antibody
Excessive conjugation may impair antigen recognition
Insufficient conjugation may result in inadequate signal
Validation for specific applications:
Isotype and Fc considerations:
Select appropriate isotype based on application needs:
For detection-only applications, F(ab')2 fragments may reduce background
The comprehensive consideration of these factors ensures selection of biotin-conjugated TNFRSF25 antibodies optimally suited for specific research objectives, enhancing experimental rigor and reproducibility.
Current research findings on TNFRSF25 provide valuable insights that will shape future biotin-conjugated antibody development and applications:
Selective T cell subset modulation:
Current understanding: TNFRSF25 and TNFRSF4 (OX40) have divergent effects on different T cell subsets
Future direction: Development of biotin-conjugated antibodies with enhanced subset selectivity
Engineer antibodies targeting specific TNFRSF25 epitopes that preferentially activate conventional T cells over Tregs
Design biotin conjugation strategies that preserve this selective activity
Structural determinants of agonistic activity:
Current knowledge: Epitope specificity critically influences agonistic potential
Future approach: Structure-guided antibody engineering
Use structural biology insights to design biotin conjugation sites that preserve critical agonistic epitopes
Develop site-specific biotinylation methods that maintain optimal receptor clustering properties
FcγR engagement requirements:
Current finding: TNFRSF25 antibody function requires engagement of both inhibitory FcγRIIB and activating FcγRIII
Future development: Optimized Fc engineering
Design antibodies with modified Fc regions to enhance FcγR engagement
Develop biotin conjugation methods that preserve critical Fc-FcγR interactions
Alternative TNFRSF25 targeting strategies:
Current research: Multimeric TL1A fusion proteins function as TNFRSF25 agonists without requiring Fc-FcγR interactions
Future direction: Multifunctional biotin-conjugated biologics
Develop biotin-conjugated multimeric TL1A constructs for enhanced targeting flexibility
Create hybrid molecules combining antibody specificity with TL1A functional domains
Combination immunotherapy potential:
Current finding: TNFRSF25 agonists induce long-term antitumor immune memory
Future application: Rational combination design
Develop optimized biotin-conjugated antibodies specifically designed for combination with checkpoint inhibitors
Create dual-targeting constructs leveraging biotin-streptavidin interactions