TNFRSF1A antibodies are monoclonal or polyclonal immunoglobulins designed to bind specifically to TNFRSF1A, a transmembrane receptor that interacts with tumor necrosis factor-alpha (TNF-α). These antibodies are used to:
Detect TNFRSF1A expression in cells or tissues via flow cytometry, immunohistochemistry (IHC), or Western blot .
Neutralize TNF-α-induced cytotoxicity by blocking receptor-ligand interactions .
Study mutations in TNFRSF1A linked to diseases like TRAPS (TNF receptor-associated periodic syndrome) and cancer .
A comparison of widely used TNFRSF1A antibodies is provided below:
Renal Cell Carcinoma (RCC): TNFRSF1A knockdown via siRNA reduced proliferation (by 40–60%), migration, and invasion in 786-O cells, highlighting its role as a therapeutic target .
Gliomas: High TNFRSF1A expression correlates with WHO grade, IDH mutation status, and poor prognosis. Antibody-mediated inhibition suppressed glioma cell proliferation and migration in vitro .
Mechanistic Insights: TNFRSF1A activates MAPK signaling, promoting tumor growth and immune evasion .
TRAPS: Mutations (e.g., p.C125Y) impair receptor shedding, leading to uncontrolled inflammation. Antibodies help quantify TNFRSF1A surface expression in monocytes .
Schizophrenia/Dementia: Elevated serum TNFRSF1A levels correlate with severe psychosis and cognitive decline .
Diagnostic Utility: TNFRSF1A antibodies enable stratification of glioma subtypes and monitoring of TRAPS progression .
Therapeutic Potential: Neutralizing antibodies (e.g., MAB225) disrupt TNF-α signaling, offering avenues for cancer immunotherapy .
Specificity: Antibodies like MAB225 show no cross-reactivity with TNFRSF1B or other TNF receptors .
Experimental Workflows:
TNFRSF1A is a cell surface receptor for tumor necrosis factor-alpha (TNF-α) that plays critical roles in inflammatory signaling, cell survival, and apoptosis. Research significance stems from its involvement in various pathways including mTOR, JNK, IKK, caspase 3, MAPK, and NF-κB signaling . The receptor contains an extracellular domain that can be proteolytically cleaved, creating soluble forms that modulate TNF signaling. TNFRSF1A has been identified as a potential biomarker in liver injury and sepsis , and it represents a therapeutic target for osteoporosis, chronic inflammatory conditions, and autoimmune diseases . Recent research has also identified TNFRSF1A as a cancer marker in renal cell carcinoma .
TNFRSF1A antibodies have been validated for multiple research applications:
Validated applications: Western Blot, Flow Cytometry, Immunocytochemistry, Immunohistochemistry, CyTOF, Agonist Activity assays
Additional cited applications: ELISA Development, Functional Assays, Immunohistochemistry (both frozen and paraffin-embedded samples), Immunoprecipitation, and Neutralization assays
When selecting an antibody, researchers should confirm validation for their specific application, as performance can vary significantly between applications even with the same antibody.
Polyclonal TNFRSF1A antibodies (like goat polyclonal IgG) recognize multiple epitopes on the target protein, potentially offering higher sensitivity but lower specificity . They are particularly useful for detecting proteins expressed at low levels or when protein conformation may be altered.
Monoclonal antibodies (such as clone #16805) recognize a single epitope, providing higher specificity but potentially lower sensitivity . They are preferable for applications requiring consistent lot-to-lot reproducibility and when distinguishing between closely related proteins.
For functional studies examining TNFRSF1A signaling, monoclonal antibodies with known epitope binding regions may be advantageous, especially when targeting specific functional domains or when agonistic/antagonistic activities are required .
Validation for Western blot applications should follow this methodological approach:
Positive controls: Include recombinant human TNFRSF1A protein (such as catalog #636-R1) as a positive control
Reduction conditions: Test under both reducing and non-reducing conditions, noting that some antibodies (e.g., clone #16805) may only detect TNFRSF1A under non-reducing conditions
Expected molecular weight: Look for a band at approximately 55 kDa, but be aware that multiple bands may appear due to:
Loading control: Include appropriate loading controls (e.g., β-Actin) to normalize protein quantities
Concentration optimization: For polyclonal antibodies, start with 0.1 μg/mL concentration and adjust as needed based on signal-to-noise ratio
A properly validated Western blot protocol should demonstrate specificity through absence of signal in negative controls and appropriate molecular weight detection in positive controls.
For optimal flow cytometric detection of TNFRSF1A:
Antibody concentration: Use approximately 2.5 μg per 10^6 cells as a starting point
Cell preparation: For human samples, peripheral blood monocytes or lymphocytes are appropriate cell types for TNFRSF1A detection
Fixation: Use mild fixation protocols; harsh fixation may destroy or mask the epitope
Controls: Include:
Isotype controls to assess non-specific binding
Unstained controls for autofluorescence calibration
FMO (fluorescence minus one) controls for accurate gating
Compensation: Proper compensation is essential when using multiple fluorophores
Permeabilization: Generally unnecessary for TNFRSF1A detection as it's primarily a cell surface receptor, but may be required for certain experimental designs
Researchers should validate protocols using cells known to express TNFRSF1A (such as activated T cells or monocytes) before proceeding with experimental samples.
For optimal immunocytochemistry detection of TNFRSF1A:
Antibody concentration: Start with 5-15 μg/mL for polyclonal antibodies
Fixation method: Use immersion fixation for suspension cells like peripheral blood lymphocytes
Incubation conditions: Typical protocol includes:
Primary antibody incubation for 3 hours at room temperature or overnight at 4°C
Secondary antibody incubation for 1-2 hours at room temperature
Counterstaining: Consider nuclear counterstaining (e.g., DAPI) for better visualization of cellular localization
Controls: Include:
Negative controls (isotype or secondary-only)
Positive controls (cells known to express TNFRSF1A)
Signal enhancement: If needed, consider tyramide signal amplification or similar methods for weak signals
For non-adherent cells, follow specialized protocols such as the "Fluorescent ICC Staining of Non-adherent Cells" protocol referenced in the literature .
When facing conflicting results across different applications:
Epitope accessibility: Different applications expose different epitopes. For example, denaturation during Western blotting may destroy conformational epitopes recognized in flow cytometry. Some antibodies (e.g., clone #16805) work under non-reducing conditions only .
Expression levels: Flow cytometry may detect surface expression while Western blot measures total protein. Discrepancies may reflect differences in localization rather than total expression.
Cross-reactivity: Some antibodies show cross-reactivity with related proteins or across species. For example, anti-human TNFRSF1A antibodies may show varying degrees of cross-reactivity with mouse TNFRSF1A .
Methodological validation: Confirm that each technique has been properly validated:
For Western blots: confirm protein size and specificity
For flow cytometry: verify with appropriate controls
For immunohistochemistry: include relevant positive and negative controls
Antibody quality: Consider antibody degradation or lot-to-lot variability
When possible, validate findings using multiple antibodies targeting different epitopes of TNFRSF1A to increase confidence in your results.
Cross-reactivity with other species occurs for several methodological reasons:
Epitope conservation: The peptide sequence corresponding to AA 20-43 of mouse TNF-R1 is identical to rat and human sequences in those residues, explaining why some antibodies show broad cross-reactivity .
Functional homology: Some antibodies recognize functional domains that are conserved across species due to evolutionary pressure to maintain receptor function.
Variable cross-reactivity by application: An antibody may demonstrate cross-reactivity in one application but not another. For example, anti-human TNFRSF1A antibodies exhibit TNF agonist activities on mouse L-929 fibroblast cells despite limited cross-reactivity in direct ELISAs (less than 20%) .
Testing methodology: Cross-reactivity should be systematically evaluated through:
Direct ELISAs with recombinant proteins from different species
Functional assays on cell lines from different species
Sequence alignment analysis of the immunogen across species
When cross-reactivity is observed, researchers should determine whether it results from specific binding to conserved domains or non-specific interactions before utilizing the antibody in cross-species experiments.
Several methodological factors influence reproducibility in functional assays:
Antibody concentration: The effective dose (ED₅₀) for agonist activity in cytotoxicity assays is typically 1-6 μg/mL, but this can vary between experiments and cell lines .
Cell line sensitivity: Different cell lines show variable sensitivity to TNFRSF1A-mediated effects. L-929 mouse fibrosarcoma cells are commonly used but require the metabolic inhibitor actinomycin D (1 μg/mL) for optimal sensitivity .
Recombinant protein quality: When using recombinant TNFRSF1A (0.3 μg/mL) and TNF-alpha (0.25 ng/mL) in neutralization assays, protein quality and activity can significantly impact results .
Incubation conditions: Temperature, duration, and media composition affect receptor-ligand interactions and downstream signaling.
Cell passage number: High passage cells may show altered receptor expression or signaling capabilities.
Readout methodology: Different viability or apoptosis assays have varying sensitivities and can produce inconsistent results.
To maximize reproducibility, researchers should standardize all experimental parameters, use low-passage cells, include appropriate controls, and perform multiple biological replicates.
For receptor neutralization and signaling studies:
Neutralization approach:
Pre-incubate cells with anti-TNFRSF1A antibody (1-10 μg/mL) before TNF-alpha stimulation
Confirm neutralization by measuring downstream signaling events such as NF-κB phosphorylation
A dose-dependent inhibition curve should demonstrate specificity (as shown in studies where increasing concentrations of TNFR1AB (4, 6, and 10 μg/mL) inhibited TNF-alpha-induced phosphorylation of p105 and p65 in a dose-dependent manner)
Signaling pathway analysis:
Functional consequences:
This approach allows detailed mapping of TNFRSF1A-dependent signaling cascades and their functional outcomes in various cellular contexts.
For single-cell analysis of TNFRSF1A:
Mass cytometry (CyTOF) applications:
Flow cytometry considerations:
Maintain consistent compensation settings across experiments
Assess TNFRSF1A expression heterogeneity within cell populations
Consider index sorting for linking protein expression to single-cell transcriptomics
Single-cell RNA-seq correlation:
Validate antibody specificity before correlating protein expression with transcriptomic data
Consider that mRNA and protein levels may not directly correlate due to post-transcriptional regulation
Recent research on renal cell carcinoma utilized single-cell approaches to examine TNFRSF1A expression heterogeneity
Spatial analysis:
Use validated immunofluorescence protocols to maintain epitope integrity
Consider multiplexing with other markers to identify specific cell populations expressing TNFRSF1A
These advanced techniques allow researchers to understand the heterogeneity of TNFRSF1A expression and its functional consequences at the single-cell level.
To investigate TNFRSF1A shedding and soluble receptor dynamics:
Detection of membrane-bound versus soluble forms:
Select antibodies that recognize either the extracellular domain (for both forms) or intracellular domain (for membrane-bound only)
Use Western blot to distinguish between the full-length (~55 kDa) and cleaved soluble forms
Different antibodies may preferentially detect specific forms or oligomerization states
Shedding pathway analysis:
Functional implications:
This approach helps elucidate the physiological and pathological roles of TNFRSF1A shedding in inflammatory regulation and disease progression.
Based on current research on TNFRSF1A as a biomarker in sepsis-related liver injury :
Tissue-specific analysis:
Compare TNFRSF1A expression across multiple tissues (liver, lung, kidney) to establish specificity
Use Western blot with anti-TNFRSF1A antibody and β-Actin as loading control
Correlate protein expression with histopathological changes (vacuolar degeneration, nuclear rupture)
Animal models:
Establish appropriate sepsis models (e.g., cecal ligation and puncture)
Include time-course analysis to determine dynamic changes in TNFRSF1A expression
Consider comparing wildtype and TNFRSF1A-/- mice to assess functional significance
Pathway investigation:
Examine relationships between TNFRSF1A and other signaling cascades:
mTOR and MAPK pathways (inflammatory regulation)
Caspase 3 and NF-κB pathways (apoptosis control)
Use specific antibodies against phosphorylated pathway components
Clinical correlation:
Translate findings to human samples when possible
Correlate TNFRSF1A levels with clinical parameters of liver dysfunction
Consider measuring soluble TNFRSF1A in patient serum/plasma
This comprehensive approach can validate TNFRSF1A as a specific biomarker for septic liver injury and elucidate underlying mechanisms.
For cancer research focused on TNFRSF1A :
Single-cell heterogeneity analysis:
Apply single-cell RNA-seq and proteomics to analyze TNFRSF1A expression variability within tumors
Use validated antibodies for flow cytometry and immunohistochemistry to map expression at the protein level
Correlate with clinical parameters and patient outcomes
Functional validation studies:
Design in vitro experiments to investigate cancer-promoting characteristics:
Proliferation assays following TNFRSF1A modulation
Migration and invasion assays
Apoptosis resistance measurements
Consider both antibody-based neutralization and genetic approaches (siRNA, CRISPR)
Signaling pathway investigation:
Examine TNFRSF1A-mediated activation of:
NF-κB pathway (survival signaling)
MAPK pathway (proliferation)
Caspase cascade (apoptosis resistance)
Use phospho-specific antibodies for key pathway components
Tumor microenvironment interaction:
Study how TNFRSF1A expression affects tumor-immune cell interactions
Investigate paracrine signaling between different cell populations
Consider multiplexed immunofluorescence or mass cytometry approaches
Particularly for renal cell carcinoma, both single-cell heterogeneity analysis and functional validation experiments have been employed to establish TNFRSF1A as a cancer marker .
To investigate TNFRSF1A as a therapeutic target :
Inhibition strategy development:
In vitro disease models:
Establish relevant cell culture systems (e.g., synoviocytes for arthritis, hepatocytes for liver inflammation)
Measure inflammatory mediators following TNFRSF1A modulation
Assess functional outcomes such as:
Cell viability and proliferation
Inflammatory cytokine production
Tissue-specific functional parameters
Selectivity assessment:
Compare effects of TNFRSF1A targeting versus other TNF receptors
Investigate potential off-target effects on related signaling pathways
Consider the balance between therapeutic efficacy and adverse effects
Translational potential evaluation:
Assess pharmacokinetics and tissue distribution of antibody-based therapeutics
Determine optimal dosing regimens for sustained pathway modulation
Consider combination approaches with existing therapies
This systematic approach can identify the most promising strategies for targeting TNFRSF1A in conditions like osteoporosis, chronic inflammatory disorders, and autoimmune diseases .