TNFRSF25 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Structure

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 .

Biochemical Properties

ParameterDetails
Host SpeciesRabbit, Goat, or Mouse
ImmunogenSynthetic peptide derived from human TNFRSF25 (residues 25–100/417)
ConjugationBiotin
ReactivityHuman, Mouse, Rat, Bovine
ApplicationsWB, ELISA, IHC-P, IHC-F, Flow Cytometry
Concentration1 µg/µL (typical)
Storage-20°C in glycerol-containing buffer

Target Details

  • Gene ID: 8718

  • Synonyms: DR3, LARD, TRAMP, Apo-3, TNFRSF12

  • Function: Regulates lymphocyte homeostasis, apoptosis, and NF-κB signaling .

Inflammatory Diseases

  • 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 .

Cancer Immunotherapy

  • 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 .

Performance in Assays

ApplicationDilution RangeKey Findings
Western Blot1:300–1:5000Detects TNFRSF25 at ~45–50 kDa in human and mouse lymphoid tissues .
IHC-P1:200–1:400Localizes TNFRSF25 in human thymocytes and inflamed intestinal epithelium .
Flow Cytometry1:100–1:500Identifies TNFRSF25+ regulatory T cells (Tregs) and activated CD4+ T cells .

Cross-Reactivity

  • Mouse DR3: Detected with ~5% cross-reactivity to human DR3 in WB .

  • Biotin Compatibility: Compatible with streptavidin-HRP or fluorophore systems for signal amplification .

Clinical and Preclinical Insights

  • 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 .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery timelines, please consult your local distributor.
Synonyms
Apo 3 antibody; Apo-3 antibody; Apo3 antibody; Apoptosis inducing receptor AIR antibody; Apoptosis inducing receptor antibody; Apoptosis mediating receptor antibody; Apoptosis mediating receptor DR 3 antibody; Apoptosis mediating receptor DR3 antibody; Apoptosis mediating receptor TRAMP antibody; Apoptosis-inducing receptor AIR antibody; Apoptosis-mediating receptor DR3 antibody; Apoptosis-mediating receptor TRAMP antibody; DDR 3 antibody; DDR3 antibody; Death domain receptor 3 antibody; Death domain receptor 3 soluble form antibody; Death receptor 3 antibody; Death receptor beta antibody; DR 3 antibody; DR3 antibody; LARD antibody; Lymphocyte associated receptor of death antibody; Lymphocyte-associated receptor of death antibody; Protein WSL antibody; Protein WSL-1 antibody; TNF receptor superfamily member 25 antibody; TNFR25 antibody; TNFRSF 12 antibody; TNFRSF 25 antibody; TNFRSF12 antibody; TNFRSF12, formerly antibody; TNFRSF25 antibody; TNR25_HUMAN antibody; TR 3 antibody; TR3 antibody; TRAMP antibody; Translocating chain association membrane protein antibody; Tumor necrosis factor receptor superfamily member 12 antibody; Tumor necrosis factor receptor superfamily member 25 antibody; Tumor necrosis factor receptor superfamily, member 12 (translocating chain association membrane protein) antibody; Tumor necrosis factor receptor superfamily, member 12, formerly antibody; WSL 1 antibody; WSL antibody; WSL LR antibody; WSL protein antibody; WSL1 antibody; WSL1 protein antibody; WSLLR antibody
Target Names
TNFRSF25
Uniprot No.

Target Background

Function
TNFRSF25, also known as Death Receptor 3 (DR3), is a receptor for TNFSF12/APO3L/TWEAK. It interacts directly with the adapter protein TRADD and plays a crucial role in mediating the activation of NF-kappa-B and inducing apoptosis. DR3 is believed to be involved in regulating lymphocyte homeostasis.
Gene References Into Functions
  1. Hypermethylation of the proapoptotic genes BCL2 L11 and TNFRSF25 has been observed in pleomorphic adenoma of the salivary glands. However, this phenomenon did not affect mRNA transcription. PMID: 28941993
  2. The DR3/TL1A pathway directly enhances human osteoclast formation and resorptive activity, controlling the expression and activation of CCL3 and MMP-9. PMID: 28062298
  3. Untreated children with inflammatory bowel disease (IBD) exhibit a higher percentage of DR3(+) peripheral blood mononuclear cells (PBMCs). PMID: 27001939
  4. In addition to apoptosis, DR3 can robustly trigger necroptotic cell death. Studies provide evidence for TL1A-induced, DR3-mediated necrosome assembly. This DR3-mediated necroptosis critically relies on receptor-interacting protein 1 (RIP1) and RIP3, the core components of the necroptotic machinery, which activate the pseudo-kinase mixed lineage kinase domain-like, the prototypic downstream effector molecule of necroptosis. PMID: 27592300
  5. Research suggests that human regulatory T-lymphocytes express DR3 and demonstrate DR3/TL1A-mediated activation of signaling via MAP kinases and NFkappaB. (DR3 = death receptor 3; TL1A/TNFSF15 = tumor necrosis factor [ligand] superfamily, member 15) PMID: 28337757
  6. These findings raise the possibility of involvement of TL1A/DR3/DR3-mediated mechanisms in epithelial-mesenchymal interactions and the development of inflammation-induced intestinal fibrosis in Crohn's disease. PMID: 27665176
  7. Activation of DR3 is accompanied by inhibition of apoptosis of naïve T-lymphocytes in children with acute infectious mononucleosis. PMID: 27682848
  8. Biologics beyond TNF-alpha inhibitors and the effect of targeting the homologues TL1A-DR3 pathway in chronic inflammatory disorders have been investigated. PMID: 26810853
  9. Higher DR3 levels have been associated with early stage chronic lymphocytic leukemia. PMID: 26393680
  10. DR3 is effectively activated by soluble TL1A trimers. PMID: 26509650
  11. These data identify new roles for DR3 in regulating osteoblast-dependent bone mineral apposition. PMID: 26065008
  12. Human memory IL-18Ralpha and DR3 CD4+ T cells may contribute to antigen-independent innate responses at barrier surfaces. PMID: 25269704
  13. DR3 is expressed by IL-22-producing human group 3 innate lymphoid cells (ILC3s). PMID: 26046454
  14. These results suggest that TL1A could promote Th17 differentiation in rheumatoid arthritis via the activation of RORc, and this effect may be mediated by the binding of TL1A with DR3. PMID: 24832108
  15. Silencing of the DR3 gene affects levels of apoptosis antigen 3 ligand in cells. PMID: 25370568
  16. DR3 is expressed in some interstitial vascular endothelial cells in human kidney in situ. These endothelial cells also respond to TL1A by activating NF-kappaB. Very low levels of DR3 are seen on the cell surface of HUVEC, which do not respond to TL1A. PMID: 25399326
  17. The changes in frequency of occurrence of spliced variants of DR3/LARD mRNA were directed towards modulation of apoptosis and restraint of antiviral immune response. PMID: 25929035
  18. Distinctions in occurrence of spectrums of DR3/LARD mRNA at healthy volunteers and colon cancer patients can define a different susceptibility of immunocompetent and tumor cells for apoptosis. PMID: 25509355
  19. DR-3 drives early cartilage destruction in the antigen-induced model of inflammatory arthritis through the release of CXCL1, maximizing neutrophil recruitment to the joint and leading to enhanced local production of cartilage-destroying enzymes. PMID: 25044706
  20. It is suggested that tectochrysin leads to apoptotic cell death in non-small cell lung cancer (NSCLC) cells through activation of DR3 and Fas expression via inhibition of STAT3 phosphorylation. PMID: 25083589
  21. TRAMP mice fed with 3'-diindolylmethane-supplemented diet show a much lower incidence of tumorigenesis and metastasis compared to the untreated control group. PMID: 23658110
  22. Collectively, these data suggest a complex role for DR3 in breast cancer development and progression. PMID: 23443464
  23. Both TNFRSF25 and TNFRSF4 independently and additively costimulate vaccine-induced CD8+ T cell proliferation following both primary and secondary antigen challenge. PMID: 22956587
  24. Protein expression of tumor necrosis factor (TNF)-like ligand 1A (TL1A) and death-domain receptor (DR)3 is upregulated in aged bladder tissues. PMID: 22641456
  25. Further investigations into the mechanisms by which the E-selectin-activated pathways downstream of DR3 confer a survival advantage to colon cancer cells have been conducted. PMID: 21722370
  26. It has been demonstrated that IL-32 enhances the cytotoxic effect of natural killer cells on prostate cancer cells through activation of DR3 and caspase-3. PMID: 22043900
  27. In active psoriasis, abundant immunostaining for TL1A and significant upregulation of its receptors DR3 and DcR3 have been observed. PMID: 21672030
  28. The role of TNFRSF25:TNFSF15 in disease and health has been investigated. PMID: 21153333
  29. DR3 has been critically implicated in the pathogenesis of rheumatoid arthritis. PMID: 20125169
  30. These data confirm that silencer of death domains (SODD) and death receptor 3 (DR3) are expressed in a regulated manner during renal transplant rejection, and identify DR3 as a potential inducible mediator of tubular inflammation and injury. PMID: 12875962
  31. TL1A-induced NF-kappaB activation and c-IAP2 production prevent DR3-mediated apoptosis. PMID: 12882979
  32. Death receptor 3 gene duplication has been associated with rheumatoid arthritis. PMID: 15241467
  33. TL1A and DR3 are involved in atherosclerosis through the induction of pro-inflammatory cytokines/chemokines. PMID: 15760679
  34. VEGI gene expression is subject to regulation by inflammatory cytokines. VEGI is also capable of regulating the expression of several important genes involved in angiogenesis. PMID: 16517446
  35. Results suggest that death receptor-3 activation can mediate apoptosis in osteoblasts, although its activity is highly restricted by its soluble ligand-binding isoform and possibly also by alternate survival signals. PMID: 16986165
  36. These findings suggest that caspase-10, DR-3 and IGFBP-3 are involved in apoptosis in the preeclamptic placenta. PMID: 17085968
  37. The TNFR25/TL1A pair provides an early signal for cytokine production in the lung and may therefore be a drug target in attempts to attenuate lung inflammation in asthmatics. PMID: 18411341
  38. The HLA-B8 and DR3 haplotype is associated with graft failure after renal transplantation in patients with underlying immunoglobulin A nephropathy. PMID: 19674013

Show More

Hide All

Database Links

HGNC: 11910

OMIM: 603366

KEGG: hsa:8718

UniGene: Hs.462529

Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein.; [Isoform 2]: Cell membrane; Single-pass type I membrane protein.; [Isoform 9]: Cell membrane; Single-pass type I membrane protein.; [Isoform 11]: Cell membrane; Single-pass type I membrane protein.; [Isoform 3]: Secreted.; [Isoform 4]: Secreted.; [Isoform 5]: Secreted.; [Isoform 6]: Secreted.; [Isoform 7]: Secreted.; [Isoform 8]: Secreted.; [Isoform 10]: Secreted.; [Isoform 12]: Secreted.
Tissue Specificity
Abundantly expressed in thymocytes and lymphocytes. Detected in lymphocyte-rich tissues such as thymus, colon, intestine, and spleen. Also found in the prostate.

Q&A

What is TNFRSF25 and why is it important in immunological research?

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 .

How does biotin conjugation enhance TNFRSF25 antibody functionality in research applications?

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.

What are the optimal protocols for using biotin-conjugated TNFRSF25 antibodies in flow cytometry?

For optimal flow cytometry results with biotin-conjugated TNFRSF25 antibodies, follow this methodological approach:

  • Sample preparation:

    • Isolate target cells (PBMCs, splenocytes, or cultured cells)

    • Determine appropriate cell concentration (typically 1×10^6 cells/100 μL)

    • Consider stimulation with PMA and calcium ionophore for 24 hours if enhanced TNFRSF25 expression is desired

  • 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:

    • When co-staining with other markers like CD3, use non-biotinylated antibodies with direct fluorophore conjugation

    • Set proper compensation controls for fluorescence spillover

    • Include FMO (Fluorescence Minus One) controls to set accurate gates

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.

How can biotin-conjugated TNFRSF25 antibodies be optimized for Western blot analysis?

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:

    • Load 20-50 μg of protein per lane

    • Use 10-12% SDS-PAGE gels (optimal for detecting the 45.4 kDa TNFRSF25 protein)

    • Include positive controls (lymphocytes or TNFRSF25-transfected cells)

  • 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:

    • Expect bands at ~45.4 kDa for canonical TNFRSF25

    • Additional bands may appear between 30-50 kDa due to the 12 reported isoforms

    • Post-translational modifications, particularly glycosylation, may cause band shifting

To validate results, consider testing crude cell lysates alongside membrane-enriched fractions, as TNFRSF25 is a transmembrane protein.

What methodological approaches can increase specificity when using biotin-conjugated TNFRSF25 antibodies in immunohistochemistry?

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:

    • Expect predominant staining in lymphocyte-rich areas

    • Compare with established T cell markers (CD3, CD4, CD8) on serial sections

    • Regulatory T cells should show particularly strong TNFRSF25 expression

This approach enhances specificity by addressing the common challenges of endogenous biotin and cross-reactivity in immunohistochemical applications.

How can biotin-conjugated TNFRSF25 antibodies be employed in studying T cell costimulation in vaccine development?

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:

    • Compare TNFRSF25 and TNFRSF4 (OX40) costimulation effects

    • Research indicates these receptors independently and additively costimulate vaccine-induced CD8+ T cell proliferation

    • Analyze differential effects on CD4+ T cell subsets, as TNFRSF25 agonists are strong costimulators of Treg proliferation

  • Adjuvant development protocol:

    • Combine TNFRSF25 agonists with traditional protein/adjuvant vaccines (e.g., OVA/aluminum hydroxide)

    • Monitor antibody production (IgG1, IgG2a, IgG2b)

    • Track memory T cell formation

  • 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:

ParameterTNFRSF25 CostimulationTNFRSF4 Costimulation
CD8+ T cell proliferationStrong enhancementStrong enhancement
CD4+ Tconv proliferationWeak costimulationPotent costimulation
Treg proliferationStrong costimulationWeak costimulation
IgG1 productionEnhancedMinimal effect
IgG2a productionEnhancedModerately enhanced
IgG2b productionEnhancedMinimal effect
Memory responseAffects secondary immunityAffects secondary immunity

This methodological approach provides insights into how TNFRSF25 targeting can enhance vaccine efficacy through costimulatory mechanisms .

What are the methodological considerations when using biotin-conjugated TNFRSF25 antibodies in cancer immunotherapy research?

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:

    • Compare biotin-conjugated antibodies with different epitope specificities

    • Research shows that antibody 1A6-m1 exhibits greater antitumor activity than higher affinity anti-TNFRSF25 antibodies targeting overlapping epitopes

    • Evaluate FcγR engagement requirements:

      • Evidence indicates antitumor effects require both inhibitory FcγRIIB and activating FcγRIII engagement

      • Modification of the CH1-hinge region influences therapeutic efficacy

  • 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:

    • Design rechallenge experiments to assess immune memory

    • Implement long-term survival studies

    • TNFRSF25 agonistic antibodies have been shown to induce long-term antitumor immune memory

  • Comparative analysis with TL1A-based approaches:

    • Research indicates multimeric TL1A fusion proteins can function as TNFRSF25 agonists without requiring Fc-FcγR interactions

    • Comparative evaluation methodology:

      • Assess T cell co-stimulation

      • Measure tumor growth inhibition

      • Evaluate toxicity profiles

These methodological considerations address the complexities of TNFRSF25 targeting in cancer immunotherapy and provide a framework for robust experimental design.

How can biotin-conjugated TNFRSF25 antibodies be utilized to investigate isoform-specific functions?

For investigating TNFRSF25 isoform-specific functions using biotin-conjugated antibodies, implement this advanced methodological framework:

  • Isoform identification protocol:

    • Human TNFRSF25 has up to 12 reported isoforms

    • Design isoform-specific RT-PCR primers

    • Sequence validate isoform expression in target tissues/cells

    • Correlate isoform expression with functional outcomes

  • 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:

    • TNFRSF25 undergoes glycosylation

    • Implement enzymatic deglycosylation (PNGase F, O-glycosidase)

    • Compare migration patterns before and after glycosidase treatment

    • Correlate glycosylation status with receptor function

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.

What are the common technical challenges when using biotin-conjugated TNFRSF25 antibodies in flow cytometry, and how can they be addressed?

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 TypeCommon IssueOptimization StrategyExpected Improvement
Resting T cellsLow TNFRSF25 expressionPMA/ionomycin stimulation (24h)2-5× signal increase
Regulatory T cellsVariable expression levelsCD25/FoxP3 co-stainingImproved population resolution
Memory T cellsBackground autofluorescenceUse streptavidin-APC or PEBetter signal-to-noise ratio
Activated T cellsReceptor internalizationFixed/permeabilized stainingDetection of total protein
ThymocytesHigh endogenous biotinExtensive biotin blockingReduced background

These methodological approaches ensure robust and reliable detection of TNFRSF25 across various experimental contexts .

How can researchers validate the functional activity of biotin-conjugated TNFRSF25 antibodies in agonist versus antagonist studies?

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:

    • For agonistic activity:

      • Assess expansion of splenic Tregs (known TNFRSF25 agonist effect)

      • Measure costimulation of antigen-specific T cell responses

    • For antagonistic activity:

      • Evaluate inhibition of TL1A-mediated inflammation

      • Assess reduction in antibody responses to T-dependent antigens

The following data table illustrates typical validation parameters:

Validation ParameterAgonistic ProfileAntagonistic ProfileNeutral Binding
T cell proliferation>200% of control<80% of TL1A response80-120% of control
NF-κB activation>2.5-fold increase<0.5-fold of TL1ANo significant change
Treg expansionSignificant increaseNo effect or decreaseNo significant change
Cytokine productionIL-2, IFN-γ increaseInhibition of TL1A-inducedNo significant change
Antitumor activityTumor regressionNo effect or promotionVariable effects

This comprehensive validation approach ensures accurate classification of biotin-conjugated TNFRSF25 antibodies as agonists or antagonists, critical for their application in immunotherapy research .

What emerging methodologies are being developed to enhance the utility of biotin-conjugated TNFRSF25 antibodies in single-cell analysis?

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.

How might biotin-conjugated TNFRSF25 antibodies contribute to developing novel combination immunotherapies?

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:

    • Methodology:

      • Prime with cancer vaccine (peptide or DNA-based)

      • Boost immune response with biotin-conjugated TNFRSF25 agonistic antibodies

    • Research basis: TNFRSF25 stimulation enhances vaccine-induced T cell responses

    • Expected outcomes:

      • Increased magnitude of antigen-specific T cell responses

      • Enhanced memory T cell formation

      • Improved antibody production (IgG1, IgG2a, IgG2b)

The following table summarizes potential combination strategies and their predicted outcomes:

Combination StrategyTarget Cell PopulationExpected MechanismPredicted Clinical Benefit
TNFRSF25 + PD-1 blockadeExhausted tumor-infiltrating T cellsRestoration of effector functionImproved response in checkpoint-resistant tumors
TNFRSF25 + TL1A fusion proteinsConventional and memory T cellsSynergistic costimulationEnhanced antitumor immunity without Fc-FcγR dependency
TNFRSF25 + radiotherapyTumor-draining lymph nodesEnhanced cross-presentationAbscopal effects on distant metastases
TNFRSF25 + CAR-T therapyEngineered T cellsSustained activation and persistenceImproved durability of response
TNFRSF25 + cancer vaccinesAntigen-specific T cellsEnhanced primary and memory responsesImproved long-term protection

These innovative approaches position biotin-conjugated TNFRSF25 antibodies as valuable tools in the development of next-generation combination immunotherapies with potentially superior efficacy profiles .

What are the key methodological considerations researchers should prioritize when selecting biotin-conjugated TNFRSF25 antibodies for their studies?

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:

    • Consider species cross-reactivity requirements:

      • Human and mouse orthologs differ significantly

      • Some antibodies (like clone 59204) recognize both human and mouse TNFRSF25

    • Match antibody species reactivity with experimental models

  • 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:

    • Confirm validation data for intended applications:

      • Flow cytometry may require different antibody characteristics than Western blotting

      • Functional assays demand validated agonistic/antagonistic properties

    • Review literature citations for application-specific performance

  • Isotype and Fc considerations:

    • Select appropriate isotype based on application needs:

      • Research demonstrates that TNFRSF25 antibody function requires engagement of both inhibitory FcγRIIB and activating FcγRIII

      • Modification of the CH1-hinge region influences therapeutic efficacy

    • 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.

How do current research findings on TNFRSF25 inform future directions for biotin-conjugated antibody development and application?

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

      • TNFRSF25 agonists strongly stimulate Treg proliferation but weakly affect conventional CD4+ T cells

      • TNFRSF4 agonists potently stimulate conventional CD4+ T cells but have minimal effect on Tregs

    • 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

      • Research demonstrates that antibody 1A6-m1 exhibits greater antitumor activity than higher affinity antibodies targeting overlapping epitopes

    • 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

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.