TNFRSF1A Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Function of TNFRSF1A Antibodies

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 .

Key Antibody Clones and Applications

A comparison of widely used TNFRSF1A antibodies is provided below:

CloneHost SpeciesApplicationsKey Findings
16803RMouseFlow cytometry, neutralization assaysInhibits TNF-α-induced cytotoxicity in L-929 fibroblasts (ND₅₀: 0.008–0.04 µg/mL) .
MAB225MouseIHC, Western blot, functional studiesReduces TNF-α-mediated NF-κB activation and SSEA-4 expression in dental pulp cells .
MAB625HumanizedELISA, neutralizationBlocks TNF-α signaling in gliomas, suppressing proliferation and migration .

Cancer Biology

  • 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 .

Inflammatory and Neurological Disorders

  • 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 .

Clinical and Therapeutic Implications

  • 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 .

Technical Considerations

  • Specificity: Antibodies like MAB225 show no cross-reactivity with TNFRSF1B or other TNF receptors .

  • Experimental Workflows:

    1. Flow Cytometry: Staining of human monocytes with anti-TNFRSF1A antibodies (clone 16803R) and APC-conjugated secondary antibodies .

    2. Neutralization Assays: Pre-incubation of TNFRSF1A antibodies (1–10 µg/mL) with recombinant TNF-α blocks cytotoxicity in L-929 cells .

Challenges and Future Directions

  • Disease Heterogeneity: Variability in TNFRSF1A shedding efficiency complicates TRAPS diagnosis .

  • Combination Therapies: Co-targeting TNFRSF1A and immune checkpoints (e.g., PD-1) may enhance anti-tumor responses .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timeframes.
Synonyms
CD120a antibody; FPF antibody; MGC19588 antibody; p55 antibody; p55-R antibody; p60 antibody; TBP1 antibody; TBPI antibody; TNF R antibody; TNF R55 antibody; TNF-R1 antibody; TNF-RI antibody; TNFAR antibody; TNFR-I antibody; TNFR1 antibody; TNFR55 antibody; TNFR60 antibody; TNFRI antibody; TNFRSF1a antibody; TNR1A_HUMAN antibody; Tumor necrosis factor receptor 1 antibody; Tumor necrosis factor receptor superfamily, member 1A antibody; Tumor necrosis factor receptor type 1 antibody; Tumor necrosis factor receptor type I antibody; Tumor necrosis factor-binding protein 1 antibody
Target Names
Uniprot No.

Target Background

Function
TNFRSF1A acts as a receptor for TNFSF2/TNF-alpha and the homotrimeric TNFSF1/lymphotoxin-alpha. The adapter molecule FADD recruits caspase-8 to the activated receptor. The resulting death-inducing signaling complex (DISC) activates caspase-8 proteolytically, initiating a cascade of caspases (aspartate-specific cysteine proteases) that ultimately lead to apoptosis. TNFRSF1A contributes to the induction of non-cytocidal TNF effects, including the establishment of an antiviral state and the activation of acid sphingomyelinase.
Gene References Into Functions
  1. Elevated TNFRs levels were associated with the risk of cardiovascular and/or all-cause mortality, independent of all relevant covariates, in patients undergoing hemodialysis. PMID: 28256549
  2. The molecular genetic marker +36G TNFR1 (OR=1,25) has been demonstrated to be involved in the development of Essential Hypertension in individuals with Metabolic Syndrome. PMID: 30289218
  3. The R426L mutation, likely de novo and novel in the TNFRSF1A gene, was not inherited from the parents, and could potentially trigger TRAPS or TRAPS-like symptoms. PMID: 27793577
  4. This study evaluated the association between TNFR1 -609G/T polymorphisms and RA susceptibility in a sample of Mexican patients. The findings suggest that the TNFR1 -609G/T polymorphisms are not associated with RA susceptibility in this population. PMID: 29404828
  5. Polymorphisms in the TNFR1 gene may have an impact on the symptomatology of schizophrenia in men. The SNPs rs4149577 and rs1860545 were associated with the intensity of the Positive and Negative Syndrome Scale (PANSS) excitement symptoms in men, potentially contributing to the risk of violent behavior. PMID: 29317797
  6. Polymorphism of the Promoter Region of the TNFRSF1A Gene is associated with Radiotherapy Induced Oral Mucositis in Head and Neck Cancer. PMID: 28401452
  7. Five single nucleotide polymorphisms in the TNFRSF1A gene were not associated with autoimmune thyroid diseases in the Chinese Han population, but rs4149570 showed a weak association with Hashimoto's thyroiditis after adjusting for gender and age. PMID: 29401539
  8. Genotype rs767455 was associated with the susceptibility of ankylosing spondylitis (AS), with the G allele of rs767455 exhibiting an association with the risk of developing AS. Only rs1061622 was significantly associated with long-term efficacy of etanercept. These findings suggest that TNFRSF1A and TNFRSF1B polymorphisms were associated with susceptibility, severity, and the long-term therapeutic efficacy of etanercept in AS patients. PMID: 30075559
  9. RACK1 associates with MOAP-1 via electrostatic associations similar to those observed between MOAP-1/RASSF1A and MOAP-1/TNF-R1. These events illustrate the complex nature of MOAP-1 regulation and highlight the important role of the scaffolding protein, RACK1, in influencing MOAP-1 biology. PMID: 29470995
  10. Serum level did not decrease significantly after tonsillectomy with steroid pulse therapy in IgA nephropathy. PMID: 28389814
  11. The TNFRSF1A c.625+10 G allele was associated with late response to anti-TNFalpha therapy, but TNFRSF1A gene SNPs is not associated with spondyloarthritis. PMID: 29579081
  12. TNFR1 is associated longitudinally with kidney function decline but not with myocardial infarct, heart failure, or mortality risk after adjustment. PMID: 28601698
  13. One third of our childhood MS patients had a heterozygous mutation in the TNFRSF1A and/or MEFV gene. This proportion significantly exceeds the number of mutations expected and was higher than in adult MS patients, suggesting that these mutations might contribute to the pathogenesis of childhood MS. PMID: 28927886
  14. This study investigated the association of NLR family pyrin domain containing 3 (NLRP3) and tumor necrosis factor receptor superfamily member 1A (TNFRSF1A) polymorphisms and haplotypes in patients with ankylosing spondylitis (AS) and treatment response to etanercept. PMID: 28116820
  15. Investigations of the underlying molecular mechanisms of TNFR1 signaling showed that PDF affects TNFR1 signaling at the proapoptotic signaling pathway by upregulation of IkappaBalpha and downregulation of cFLIPL. PMID: 28096440
  16. Case Report: tumor necrosis factor receptor-associated periodic syndrome due to the R92Q TNFRSF1A variant associated with recurrent pericarditis and cardiac tamponade. PMID: 27990755
  17. Serum TNFR1 is a biomarker for patients with chronic kidney disease. PMID: 28667032
  18. This study demonstrated that TNFR1 expression levels are related to major depressive disorder and, in conjunction, mediate the effect of childhood maltreatment history on the risk of developing major depressive disorder. PMID: 28384542
  19. SNP (36A>G) is a strong risk factor for odontogenic keratocystic tumor. PMID: 28199753
  20. Data suggest that Fas and TNFR1 are involved in glaucoma mechanisms in the cornea; the pro-apoptotic effect of the anti-glaucoma medication clonidine on corneal epithelial cells triggers Fas/TNFR1-mediated, mitochondria-dependent signaling pathway. (Fas = Fas cell surface death receptor; TNFR1 = TNF receptor superfamily member 1A) PMID: 28115640
  21. These results indicate that TNFRI-Fc and hHO-1 overexpression may apparently induce free iron in the liver and exert oxidative stress by enhancing reactive oxygen species production and blocking normal postneonatal liver metabolism. PMID: 28503569
  22. Data suggest that elevated serum levels of soluble TNF receptors, especially sTNFR1, are associated with loss of kidney function in Hispanic patients with diabetes type 2 in Colombia. PMID: 27068267
  23. Case Report: heterozygous missense variants in TNFRSF1A identified in family members with familial Mediterranean fever. PMID: 29148404
  24. Case Report: autoinflammatory syndrome with relapsing aseptic neutrophilic meningitis and chronic myelitis associated with MEFV/TNFRSF1A mutations. PMID: 28134085
  25. In this article, we will review the role of ubiquitination and proteolysis in these diverse events, focusing on our own contributions to the lysosomal apoptotic pathway linked to the subcellular compartmentalization of TNF-R1. PMID: 28765050
  26. Coadministration of either ATROSAB or EHD2-scTNFR2 into the magnocellular nucleus basalis significantly protected cholinergic neurons and their cortical projections against cell death, and reversed the neurodegeneration-associated memory impairment in a passive avoidance paradigm. Simultaneous blocking of TNFR1 and TNFR2 signaling, however, abrogated the therapeutic effect. PMID: 27791020
  27. Data show that interleukin-2 receptor alpha, tumor necrosis factor receptor 1, serum STimulation-2 (IL1RL1 gene product), and regenerating islet-derived 3-alpha were significantly associated with non-relapse mortality. PMID: 28126963
  28. Report of a severe case of TRAPS associated with a novel mutation, Thr90Pro, in the TNFRSF1A gene in an infant and several family members. PMID: 28427379
  29. Atopic dermatitis patients had increased TNFR1 expression on immune cells. PMID: 29212072
  30. Elevated levels of soluble tumor necrosis factor receptors 1 and lower levels of leptin are associated with higher developmental outcomes in infants between the ages of 6 and 24 months. PMID: 28238825
  31. The highest levels of TNFR1 are independently associated with progression of renal disease and death in type 2 diabetic nephropathy. PMID: 27003829
  32. High plasma levels of TNFR1 and TNFR2 were associated with incident intracerebral hemorrhage. PMID: 28830973
  33. Renal clear cell carcinoma cells express increased amounts of RIPK1 and RIPK3 and are poised to undergo necroptosis in response to TNFR1 signaling. PMID: 27362805
  34. TRIM28 acts as a central factor in controlling endothelial inflammatory responses and angiogenic activities by retaining expression of TNFR-1 and -2 and VEGF receptor 2 in endothelial cells. PMID: 28159803
  35. A specific link between the penetrance of the TNFRSF1A mutation and the observed T cell phenotype is reported. PMID: 26598380
  36. Collectively, this study provides further insights into RELT expression, RELT family member function, and the mechanism of RELT-induced death. PMID: 28688764
  37. Burkholderia cenocepacia BcaA binds to tumor necrosis factor receptor 1. PMID: 27684048
  38. TNFRSF1A variants were identified in 10 tumor necrosis factor receptor-associated periodic syndrome patients from 10 independent families. The T61I variant was found in patients, while the V136M and S321I variants were found in 1 patient each. All the patients were heterozygous for the variants. Among the healthy controls, 7 of 363 individuals were heterozygous for the T61I variant. PMID: 27332769
  39. Voxel-based morphometry was used to analyze the associations between TNFRSF1A (rs4149576 and rs4149577) and gray matter structure. Highly significant genotypic associations with striatal volume but not the hippocampus were seen. Specifically, for rs4149576, G homozygotes were associated with reduced caudate nucleus volumes relative to A homozygotes and heterozygotes. Reduced caudate volumes were observed in C homozygotes. PMID: 27528091
  40. Circulating TNFR1 and 2 were shown to be associated with cardiovascular disease, independent of age, sex, inflammatory markers, and other cardiovascular disease risk factors, in chronic kidney disease patients. PMID: 28489742
  41. Infection with C. trachomatis disrupts TNFR1 signaling specifically at the level of receptor internalization. PMID: 27062399
  42. Data suggest that TRAF2 (TNF receptor-associated factor 2) negatively regulates (1) TNFR1- (tumor necrosis factor binding protein 1)-induced apoptosis, (2) TNFR2- (tumor necrosis factor receptor type 2)-induced non-canonical NFkappaB signaling, and (3) TNF- (tumor necrosis factor)-induced necroptosis. [REVIEW] PMID: 26993379
  43. Our analysis showed that the TNFR1 rs2234649 polymorphism does not increase ankylosing spondylitis risk. In conclusion, the TNFR1 gene polymorphism tested does not appear to be useful for assessing predisposition to ankylosing spondylitis. PMID: 28363009
  44. Elevated serum level associated with heart failure risk in type 2 diabetes mellitus patients. PMID: 28367848
  45. The data demonstrate a novel and unexpected function of BIG1 that regulates TNFR1 signaling by targeting TRAF2. PMID: 27834853
  46. Data indicate that plasma concentrations of TNFR1 and TNFR2 are elevated in pediatric lupus nephritis. PMID: 26854079
  47. TNFR1 is the major pro-inflammatory mediator of TNF-alpha in fibroblast-like synoviocytes (FLS), whereas TNFR2 may act as an immunosuppressor in FLS for the prevention of overwhelming inflammatory reactions. PMID: 28150360
  48. Results suggest that miR-29a is an important regulator of tumor necrosis factor receptor 1 expression in breast cancer and functions as a tumor suppressor by targeting tumor necrosis factor receptor 1 to influence the growth of MCF-7 cell. PMID: 28222663
  49. Serum sTNFR1 and sTNFR2 are associated with obese girls but not obese boys, suggesting that serum sTNFRs in early childhood obesity may be sex-related. PMID: 27040725
  50. SCCAg, CYFRA 21.1, IL-6, VEGF, and sTNF receptors have roles in squamous cell cervical cancer. PMID: 26289850

Show More

Hide All

Database Links

HGNC: 11916

OMIM: 142680

KEGG: hsa:7132

STRING: 9606.ENSP00000162749

UniGene: Hs.279594

Involvement In Disease
Familial hibernian fever (FHF); Multiple sclerosis 5 (MS5)
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Golgi apparatus membrane; Single-pass type I membrane protein. Secreted. Note=A secreted form is produced through proteolytic processing.; [Isoform 4]: Secreted. Note=Lacks a Golgi-retention motif, is not membrane bound and therefore is secreted.

Q&A

What is TNFRSF1A and why is it significant in research?

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 .

What are the validated applications for TNFRSF1A antibodies?

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.

How do polyclonal and monoclonal TNFRSF1A antibodies differ in research applications?

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 .

How should TNFRSF1A antibodies be validated for Western blot applications?

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:

    • Oligomerization

    • Self-aggregation

    • Cleavage of the TNF-R1 extracellular domain

  • 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.

What are the optimal conditions for flow cytometric detection of TNFRSF1A?

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.

How can I optimize immunocytochemistry protocols for TNFRSF1A detection?

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 .

How do I interpret conflicting TNFRSF1A antibody results between different applications?

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.

Why might TNFRSF1A antibodies show unexpected cross-reactivity with other species?

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.

What factors affect the reproducibility of functional assays using TNFRSF1A antibodies?

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.

How can TNFRSF1A antibodies be used in receptor neutralization and signaling studies?

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:

    • Monitor pathway-specific readouts such as:

      • Phosphorylation of NF-κB (p65, p105) via Western blot at 5 minutes post-stimulation

      • Activation of mTOR, JNK, IKK, caspase 3, and MAPK pathways

    • Use pathway-specific inhibitors in parallel to confirm specificity

  • Functional consequences:

    • Assess cell-specific outcomes such as:

      • Cytotoxicity in sensitive cell lines like L-929

      • Expression of stemness markers (SSEA-4, OCT-4, NANOG) in stem cells

      • Inflammatory cytokine production

This approach allows detailed mapping of TNFRSF1A-dependent signaling cascades and their functional outcomes in various cellular contexts.

What are the considerations when using TNFRSF1A antibodies in single-cell analysis techniques?

For single-cell analysis of TNFRSF1A:

  • Mass cytometry (CyTOF) applications:

    • Use CyTOF-ready antibodies without BSA or carrier proteins that could interfere with metal conjugation

    • Validate panel design to avoid signal overlap between channels

    • Consider fixation-sensitive epitopes that may be affected by sample processing

  • 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.

How can TNFRSF1A antibodies be utilized in studies of receptor shedding and soluble receptor dynamics?

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:

    • Monitor the iNOS-cGMP-TACE pathway implicated in TNFR1 shedding in hepatocytes following bacterial lipopolysaccharide stimulation

    • Use pharmacological inhibitors of metalloproteinases (e.g., TAPI-1) to block shedding

    • Measure soluble TNFRSF1A in culture supernatants by ELISA

  • Functional implications:

    • Investigate how soluble TNFRSF1A modulates TNF signaling through neutralization assays

    • Examine the balance between pro-inflammatory (membrane-bound) and potentially anti-inflammatory (soluble) forms

    • Consider the role of soluble TNFRSF1A as a biomarker in conditions like sepsis-related liver injury

This approach helps elucidate the physiological and pathological roles of TNFRSF1A shedding in inflammatory regulation and disease progression.

How should researchers design experiments to investigate TNFRSF1A as a biomarker in liver injury and sepsis?

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.

What methodological approaches are recommended for studying TNFRSF1A in cancer research?

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 .

How can researchers investigate TNFRSF1A as a therapeutic target in inflammatory and autoimmune diseases?

To investigate TNFRSF1A as a therapeutic target :

  • Inhibition strategy development:

    • Use neutralizing antibodies at various concentrations (4-10 μg/mL) to block receptor function

    • Compare antibody-based approaches with soluble receptor mimetics or small molecule inhibitors

    • Assess differential effects on various signaling pathways

  • 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 .

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 2024 Thebiotek. All Rights Reserved.