TNFRSF1A Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Flow Cytometry

FITC-conjugated TNFRSF1A antibodies enable quantitative analysis of receptor expression on immune cells. For example:

  • Monocyte Studies: Staining peripheral blood monocytes with these antibodies allows detection of TNFRSF1A surface expression, as demonstrated in studies analyzing receptor shedding under PMA (phorbol ester) stimulation .

  • Gating Strategy: CD14+ monocytes are typically gated for analysis, with fluorescence intensity measured to assess receptor density changes .

Immunofluorescence (IF) and Immunohistochemistry (IHC)

These antibodies localize TNFRSF1A in tissue sections or cultured cells. For instance, they have been used to visualize receptor distribution in inflammatory diseases .

Western Blot (WB)

Although less common due to FITC’s susceptibility to denaturation, these antibodies can detect TNFRSF1A in lysates under non-reducing conditions .

Mechanistic Insights into TNFRSF1A Shedding

A 2024 study investigated the rare p.C125Y mutation in TNFRSF1A and its impact on receptor shedding in TRAPS (TNF Receptor-Associated Periodic Syndrome) patients :

  • Methodology: PBMCs from patients and controls were stimulated with PMA, and TNFRSF1A levels on CD14+ monocytes were analyzed using flow cytometry.

  • Results: The p.C125Y mutation did not impair receptor shedding, as TNFRSF1A median fluorescence intensity (MFI) decreased comparably to wild-type controls during PMA treatment .

Table 2: PMA-Induced TNFRSF1A Shedding in CD14+ Monocytes

GroupMFI Ratio (PMA/Unstimulated) at 60 MinStatistical Significance
p.C125Y Mutation (n=4)0.62 ± 0.08p > 0.05 vs. WT
Wild-Type (n=4)0.58 ± 0.06

This finding contrasts with earlier hypotheses linking TRAPS mutations to defective shedding .

Limitations and Considerations

  • Cross-Reactivity: None reported with murine or rat TNFRSF1A homologs .

  • Functional Assays: FITC conjugation does not interfere with receptor-ligand binding, making these antibodies suitable for live-cell imaging .

Future Directions

Recent advancements in multiplex flow cytometry could leverage FITC-conjugated TNFRSF1A antibodies alongside other fluorophores to study co-receptor interactions in autoimmune diseases or cancer immunotherapy contexts.

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 timelines.
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 Antibody, FITC conjugated recognizes the Tumor Necrosis Factor Receptor Superfamily Member 1A (TNFR1), a receptor for TNFSF2/TNF-alpha and homotrimeric TNFSF1/lymphotoxin-alpha. Upon activation, the adapter molecule FADD recruits caspase-8 to the receptor, forming a death-inducing signaling complex (DISC). This complex initiates caspase-8 proteolytic activation, triggering a cascade of caspases (aspartate-specific cysteine proteases) that ultimately lead to apoptosis. TNFR1 also contributes to non-cytocidal TNF effects, including the induction of an anti-viral state and activation of acid sphingomyelinase.
Gene References Into Functions
  1. Elevated TNFRs levels have been linked to an increased risk of cardiovascular and/or all-cause mortality in patients undergoing hemodialysis, independent of other relevant factors. PMID: 28256549
  2. The molecular genetic marker +36G TNFR1 (OR=1,25) has been implicated in the development of Essential Hypertension in individuals with Metabolic Syndrome. PMID: 30289218
  3. A de novo and novel mutation, R426L, in the TNFRSF1A gene has been observed, potentially triggering TRAPS or TRAPS-like symptoms. PMID: 27793577
  4. A study examining TNFR1 -609G/T polymorphisms in Mexican patients with Rheumatoid Arthritis (RA) did not find an association between these polymorphisms and RA susceptibility. PMID: 29404828
  5. Polymorphisms in the TNFR1 gene may influence the symptomatology of schizophrenia in men. SNPs rs4149577 and rs1860545 were associated with the intensity of Positive and Negative Syndrome Scale (PANSS) excitement symptoms in men, potentially contributing to the risk of violent behavior. PMID: 29317797
  6. Polymorphism in the promoter region of the TNFRSF1A gene has been associated with Radiotherapy-Induced Oral Mucositis in Head and Neck Cancer. PMID: 28401452
  7. While five single nucleotide polymorphisms in the TNFRSF1A gene were not associated with autoimmune thyroid diseases in the Chinese Han population, rs4149570 showed a weak association with Hashimoto's thyroiditis after adjusting for gender and age. PMID: 29401539
  8. Genotype rs767455 has been associated with the susceptibility of ankylosing spondylitis (AS), with the G allele showing an association with the risk of developing AS. rs1061622 was significantly associated with the long-term efficacy of etanercept. These findings suggest that TNFRSF1A and TNFRSF1B polymorphisms may be linked to the susceptibility, severity, and therapeutic response to etanercept in AS patients. PMID: 30075559
  9. RACK1 interacts with MOAP-1 through electrostatic associations, similar to those observed between MOAP-1/RASSF1A and MOAP-1/TNF-R1. This highlights the intricate regulation of MOAP-1 and the significant role of the scaffolding protein, RACK1, in influencing MOAP-1 biology. PMID: 29470995
  10. Serum levels of TNFR1 did not decrease significantly after tonsillectomy with steroid pulse therapy in patients with IgA nephropathy. PMID: 28389814
  11. The TNFRSF1A c.625+10 G allele was associated with a delayed response to anti-TNFalpha therapy, but TNFRSF1A gene SNPs were not associated with spondyloarthritis. PMID: 29579081
  12. TNFR1 has been found to be longitudinally associated with kidney function decline, but not with myocardial infarct, heart failure, or mortality risk after adjusting for relevant factors. PMID: 28601698
  13. One third of childhood MS patients exhibited a heterozygous mutation in the TNFRSF1A and/or MEFV gene. This proportion is significantly higher than expected and exceeds the number of mutations observed in adult MS patients, suggesting a potential role of these mutations in the pathogenesis of childhood MS. PMID: 28927886
  14. A 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 their treatment response to etanercept. PMID: 28116820
  15. Investigations into the underlying molecular mechanisms of TNFR1 signaling revealed that PDF affects this pathway by upregulating IkappaBalpha and downregulating cFLIPL, influencing the proapoptotic signaling pathway. PMID: 28096440
  16. A case report describes 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 has been identified as a biomarker for patients with chronic kidney disease. PMID: 28667032
  18. Research suggests that TNFR1 expression levels are linked to major depressive disorder and mediate the effect of childhood maltreatment history on the risk of developing this disorder. PMID: 28384542
  19. SNP (36A>G) has been identified as a strong risk factor for odontogenic keratocystic tumor. PMID: 28199753
  20. Data suggests that Fas and TNFR1 play a role in glaucoma mechanisms in the cornea. The pro-apoptotic effect of the anti-glaucoma medication clonidine on corneal epithelial cells triggers a Fas/TNFR1-mediated, mitochondria-dependent signaling pathway. (Fas = Fas cell surface death receptor; TNFR1 = TNF receptor superfamily member 1A). PMID: 28115640
  21. Overexpression of TNFRI-Fc and hHO-1 may induce free iron in the liver, leading to oxidative stress by enhancing reactive oxygen species production and disrupting normal postneonatal liver metabolism. PMID: 28503569
  22. Elevated serum levels of soluble TNF receptors, particularly sTNFR1, have been associated with a decline in kidney function in Hispanic patients with type 2 diabetes in Colombia. PMID: 27068267
  23. A case report identifies heterozygous missense variants in TNFRSF1A in family members with familial Mediterranean fever. PMID: 29148404
  24. A case report describes an autoinflammatory syndrome with relapsing aseptic neutrophilic meningitis and chronic myelitis associated with MEFV/TNFRSF1A mutations. PMID: 28134085
  25. This article reviews the role of ubiquitination and proteolysis in various cellular events, focusing on 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, reversing the neurodegeneration-associated memory impairment in a passive avoidance paradigm. However, simultaneous blocking of TNFR1 and TNFR2 signaling abolished the therapeutic effect. PMID: 27791020
  27. Research has shown that interleukin-2 receptor alpha, tumor necrosis factor receptor 1, serum STimulation-2 (IL1RL1 gene product), and regenerating islet-derived 3-alpha are significantly associated with non-relapse mortality. PMID: 28126963
  28. A report describes 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. Patients with atopic dermatitis have exhibited increased TNFR1 expression on immune cells. PMID: 29212072
  30. Elevated levels of soluble tumor necrosis factor receptors 1 and lower levels of leptin have been associated with better developmental outcomes in infants between 6 and 24 months of age. PMID: 28238825
  31. The highest levels of TNFR1 are independently associated with the progression of renal disease and death in type 2 diabetic nephropathy. PMID: 27003829
  32. High plasma levels of TNFR1 and TNFR2 have been linked to an increased risk of incident intracerebral hemorrhage. PMID: 28830973
  33. Renal clear cell carcinoma cells express elevated amounts of RIPK1 and RIPK3 and are prone to undergo necroptosis in response to TNFR1 signaling. PMID: 27362805
  34. TRIM28 plays a crucial role in regulating endothelial inflammatory responses and angiogenic activities by maintaining the expression of TNFR-1 and -2 and VEGF receptor 2 in endothelial cells. PMID: 28159803
  35. A study reports a specific link between the penetrance of the TNFRSF1A mutation and the observed T cell phenotype. PMID: 26598380
  36. 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 multiple patients, while the V136M and S321I variants were found in one patient each. All patients were heterozygous for the variants. Among healthy controls, 7 out 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 grey matter structure. Highly significant genotypic associations with striatal volume, but not the hippocampus, were observed. Specifically, for rs4149576, individuals with the GG genotype showed reduced caudate nucleus volumes compared to those with the AA genotype and heterozygotes. Reduced caudate volumes were also observed in individuals with the CC genotype for rs4149577. 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 patients with chronic kidney disease. 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. Analysis indicates that the TNFR1 rs2234649 polymorphism does not increase the risk of ankylosing spondylitis. Therefore, this specific TNFR1 gene polymorphism may not be useful for assessing predisposition to ankylosing spondylitis. PMID: 28363009
  44. Elevated serum levels of TNFR1 have been associated with an increased risk of heart failure in patients with type 2 diabetes mellitus. PMID: 28367848
  45. Research demonstrates a novel and unexpected function of BIG1 in regulating TNFR1 signaling by targeting TRAF2. PMID: 27834853
  46. Plasma concentrations of TNFR1 and TNFR2 have been found to be elevated in pediatric lupus nephritis. PMID: 26854079
  47. TNFR1 is the primary pro-inflammatory mediator of TNF-alpha in fibroblast-like synoviocytes (FLS), while TNFR2 may act as an immunosuppressor in FLS to prevent excessive inflammatory reactions. PMID: 28150360
  48. miR-29a has been shown to be a significant regulator of tumor necrosis factor receptor 1 expression in breast cancer, acting as a tumor suppressor by targeting tumor necrosis factor receptor 1 and influencing the growth of MCF-7 cells. PMID: 28222663
  49. Serum sTNFR1 and sTNFR2 levels are associated with obese girls but not obese boys, suggesting a potential sex-related difference in serum sTNFRs in early childhood obesity. 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 an important research target?

TNFRSF1A (Tumor Necrosis Factor Receptor Superfamily Member 1A), also known as TNF-R1, TNF-RI, TNFR-I, p55, p60, or CD120a, functions as a primary receptor for tumor necrosis factor alpha (TNF-α). This receptor plays a critical role in initiating cellular signaling pathways involved in inflammation, apoptosis, and immune regulation. TNFRSF1A has been extensively studied due to its central role in various pathological conditions including cancer, cardiovascular diseases, and inflammatory disorders. The receptor acts as a membrane-bound protein that can be cleaved into a membrane form and a soluble TNF-binding protein 1 (TBPI) . Its interaction with TNF-α initiates complex downstream signaling cascades that regulate critical cellular processes, making it a valuable target for both basic research and therapeutic development .

What are the key characteristics of FITC-conjugated TNFRSF1A antibodies?

FITC-conjugated TNFRSF1A antibodies are immunological reagents where the antibody targeting TNFRSF1A is chemically linked to fluorescein isothiocyanate (FITC), a fluorescent dye emitting green light when excited. These conjugated antibodies are available in various formats including monoclonal (derived from mouse hosts, such as clone H398 with IgG2a isotype) and polyclonal versions (typically derived from rabbit hosts) . Key characteristics include:

PropertyMonoclonalPolyclonal
Host SpeciesMouse Rabbit
ReactivityHuman, Rabbit Human
PurificationProtein A affinity chromatography Protein G (>95% purity)
Storage BufferNot specified in sources50% Glycerol, 0.01M PBS, pH 7.4 with 0.03% Proclin 300
Recommended StorageNot specified in sources-20°C or -80°C
Primary ApplicationsFlow cytometry (FACS) ELISA

These antibodies maintain their specific binding capability to TNFRSF1A while allowing direct visualization through fluorescence detection systems, making them particularly valuable for applications requiring simultaneous detection of multiple cellular markers .

What experimental applications are FITC-conjugated TNFRSF1A antibodies suitable for?

FITC-conjugated TNFRSF1A antibodies are versatile tools applicable to several experimental techniques in immunological and cell biology research. The primary applications include:

  • Flow Cytometry (FACS): The most common application, allowing quantitative analysis of TNFRSF1A expression on cell populations. These antibodies have been validated for detecting TNFRSF1A on human blood monocytes and other cell types .

  • Immunofluorescence (IF): For visualizing TNFRSF1A localization within cells or tissues using fluorescence microscopy.

  • ELISA: Some FITC-conjugated antibodies have been validated for ELISA applications, allowing quantitative measurement of TNFRSF1A in solution .

  • Immunocytochemistry (ICC): For detecting TNFRSF1A in fixed cells while preserving cellular morphology.

While Western blotting is technically possible, the FITC conjugation may not be ideal for this application as it requires additional detection methods beyond standard chemiluminescence. For experimental optimization, researchers should consider that some TNFRSF1A antibodies perform optimally under non-reducing conditions .

How should FITC-conjugated TNFRSF1A antibodies be stored and handled?

Proper storage and handling of FITC-conjugated TNFRSF1A antibodies are crucial for maintaining their performance and extending shelf-life. The recommended practices include:

  • Temperature management: Store at -20°C to -80°C for long-term preservation of antibody activity and fluorescence intensity . Avoid repeated freeze-thaw cycles as they can degrade both the antibody and the fluorophore.

  • Light protection: FITC is susceptible to photobleaching, so antibodies should be stored in amber vials or wrapped in aluminum foil to protect from light exposure. During experimental procedures, minimize exposure to direct light.

  • Buffer considerations: These antibodies are typically provided in specialized buffers (e.g., 50% Glycerol, 0.01M PBS, pH 7.4 with 0.03% Proclin 300 as preservative) . Do not alter this buffer unless absolutely necessary for your application.

  • Working solution preparation: When diluting for experiments, use fresh, high-quality buffers preferably containing 1-2% carrier protein (BSA or serum) to prevent non-specific binding and preserve activity. Calculate exact concentrations needed to minimize waste of valuable reagent.

  • Contamination prevention: Use sterile technique when handling these antibodies to prevent microbial growth that could degrade the reagent and compromise experimental results.

Maintaining a laboratory log of freeze-thaw cycles and conducting periodic quality control tests (e.g., flow cytometry with positive control samples) can help monitor antibody performance over time.

How can researchers optimize flow cytometry protocols using FITC-conjugated TNFRSF1A antibodies?

Optimizing flow cytometry protocols with FITC-conjugated TNFRSF1A antibodies requires careful consideration of several critical parameters:

  • Titration optimization: Rather than following manufacturer-suggested dilutions blindly, perform antibody titration experiments with a range of concentrations (typically 0.008-0.04 μg/mL for some clones) to determine the optimal signal-to-noise ratio for your specific cell type. Create a titration curve measuring both positive population separation and mean fluorescence intensity.

  • Compensation strategy: Since FITC has spectral overlap with other common fluorophores (particularly PE), proper compensation is essential for multiparameter analysis. Use single-stained controls with the same FITC-conjugated antibodies rather than generic FITC beads to account for fluorophore:protein ratio differences.

  • Cell preparation considerations:

    • For membrane-bound TNFRSF1A: Avoid harsh enzymatic dissociation methods that might cleave the receptor

    • For intracellular detection: Use appropriate fixation and permeabilization buffers validated for maintaining FITC fluorescence

  • Blocking strategy: Include a pre-incubation step with 5-10% serum (matching secondary antibody species if applicable) or commercial Fc blocking reagents to reduce non-specific binding, particularly important when working with samples containing Fc receptor-expressing cells like monocytes and macrophages .

  • Controls implementation:

    • Include isotype control antibody (FITC-conjugated IgG2a for monoclonal antibodies)

    • Use known TNFRSF1A-positive cell populations (human blood monocytes) as positive controls

    • Consider including a competitive binding control with unconjugated anti-TNFRSF1A

  • Data analysis refinement: Apply consistent gating strategies based on fluorescence-minus-one (FMO) controls rather than isotype controls for more accurate population identification.

What are the critical considerations when using TNFRSF1A antibodies in functional inhibition assays?

When designing functional inhibition assays using TNFRSF1A antibodies, researchers should address several considerations to ensure reliable and reproducible results:

  • Mechanism of inhibition: TNFRSF1A antibodies can inhibit TNF-α signaling through multiple mechanisms:

    • Competitive binding to prevent TNF-α interaction

    • Receptor internalization

    • Conformational changes affecting downstream signaling

  • Assay selection and optimization: The L-929 mouse fibroblast cytotoxicity assay is well-established for evaluating TNFRSF1A-TNF-α interactions . This requires:

    • Careful titration of TNF-α concentration (typically 0.25 ng/mL)

    • Co-incubation with recombinant TNFRSF1A (15 ng/mL)

    • Inclusion of metabolic inhibitors like actinomycin D (1 μg/mL)

    • Readout optimization (e.g., using Resazurin for viability assessment)

  • Neutralization curve establishment: Generate complete neutralization curves by testing antibody concentrations across a broad range (typically 0.001-1.0 μg/mL). The neutralization dose 50 (ND50) for effective antibodies typically falls between 0.008-0.04 μg/mL .

  • Species cross-reactivity considerations: Despite sequence homology, significant functional differences exist between human and mouse TNFRSF1A. Carefully validate antibodies when working across species, as many antibodies show species-specific inhibitory properties.

  • Control implementation: Include critical controls:

    • Isotype-matched control antibodies

    • Known blocking anti-TNFRSF1A antibodies (e.g., clone 16803)

    • Soluble TNFRSF1A receptor as a positive control for TNF-α inhibition

  • Readout selection: Consider multiple readouts beyond cell viability, including:

    • Signaling pathway activation (NF-κB translocation)

    • Downstream gene expression

    • Inflammatory cytokine production

This comprehensive approach enables more accurate interpretation of antibody-mediated functional effects on TNFRSF1A signaling.

How does the choice between monoclonal and polyclonal FITC-conjugated TNFRSF1A antibodies impact experimental outcomes?

The decision between monoclonal and polyclonal FITC-conjugated TNFRSF1A antibodies significantly influences experimental design, data interpretation, and research outcomes:

ParameterMonoclonal TNFRSF1A AntibodiesPolyclonal TNFRSF1A AntibodiesExperimental Impact
Epitope RecognitionSingle epitope (e.g., clone H398) Multiple epitopes across TNFRSF1A Polyclonals may provide stronger signals but with potential increased background
Lot-to-Lot ConsistencyHigh consistency between lotsVariable between lotsMonoclonals offer better reproducibility for longitudinal studies
Signal IntensityModerate (depends on epitope accessibility)Typically higher due to multiple binding sitesPolyclonals may be preferred for detecting low-abundance receptors
Background SignalGenerally lowerPotentially higherMonoclonals may provide better signal-to-noise ratio in complex samples
Receptor Conformation SensitivityMay be sensitive to conformational changesLess affected by single conformational changesConsider when studying receptor dynamics or conformation-specific interactions
TNF-α Binding InterferenceMay specifically block or not block depending on epitopeGenerally higher likelihood of functional interferenceCritical for choosing antibodies for functional studies vs. detection only
Application SuitabilityExcellent for precise epitope mapping and standardized assaysBetter for initial detection and screeningApplication-dependent selection necessary

For advanced research questions examining receptor internalization, conformational changes, or interactions with specific domains, epitope-specific monoclonal antibodies targeting defined regions of TNFRSF1A (e.g., antibodies against amino acids 20-43 versus 249-455) provide more precise mechanistic insights, though potentially at the cost of signal intensity.

What approaches can address non-specific binding and background issues with FITC-conjugated TNFRSF1A antibodies?

Non-specific binding and background fluorescence are common challenges when working with FITC-conjugated TNFRSF1A antibodies, particularly in complex biological samples. Advanced researchers can implement several strategies to mitigate these issues:

  • Sample-specific blocking optimization:

    • For peripheral blood: Use 2-5% serum matching the host species of secondary antibody

    • For tissues with high Fc receptor expression: Implement dedicated Fc receptor blocking (human TruStain FcX™ or similar)

    • For tissues with high endogenous biotin: Add avidin/biotin blocking steps

  • Advanced fixation and permeabilization protocols:

    • Membrane TNFRSF1A detection: Use mild fixation (0.5-1% paraformaldehyde) to preserve epitope structure

    • Intracellular TNFRSF1A detection: Compare commercial permeabilization kits to identify optimal conditions that maintain both epitope recognition and cellular morphology

  • Buffer optimization strategies:

    • Add 0.1-0.5% non-ionic detergents (Tween-20, Triton X-100) to reduce hydrophobic interactions

    • Incorporate electrostatically neutral proteins (1-2% BSA) to block non-specific binding sites

    • Consider adding mild reducing agents (5mM β-mercaptoethanol) if antibody performance permits

  • Advanced negative controls beyond isotype:

    • Pre-absorption controls (antibody pre-incubated with recombinant TNFRSF1A)

    • Biological negative controls (cell lines with TNFRSF1A knockout)

    • Fluorescence-minus-one (FMO) controls for multiparameter flow cytometry

  • Signal amplification considerations:

    • For very low expression: Consider tyramide signal amplification protocols adapted for FITC-conjugated antibodies

    • For multiplexing: Use sequential detection protocols with careful intermediate blocking steps

  • Image-based applications:

    • Implement computational background subtraction algorithms specific to the fluorescence profile of FITC

    • Use spectral unmixing for samples with significant autofluorescence in the FITC channel

These approaches should be systematically evaluated and optimized for each specific experimental system and biological sample type.

How can researchers validate the specificity of FITC-conjugated TNFRSF1A antibodies in their experimental systems?

Thorough validation of FITC-conjugated TNFRSF1A antibodies is essential for ensuring experimental rigor and reproducibility. A comprehensive validation strategy should include:

  • Expression system correlation verification:

    • Test antibody on cell lines with known differential TNFRSF1A expression levels

    • Compare antibody staining patterns with mRNA expression data from qPCR or RNA-seq

    • Validate with cells transfected with TNFRSF1A expression constructs versus empty vector controls

  • Genetic validation approaches:

    • Use CRISPR/Cas9 TNFRSF1A knockout cells as negative controls

    • Implement siRNA/shRNA knockdown systems with appropriate scrambled controls

    • Test on patient-derived cells with known TNFRSF1A mutations

  • Biochemical validation methods:

    • Perform competitive binding assays with increasing concentrations of recombinant TNFRSF1A protein

    • Compare staining patterns between antibodies targeting different TNFRSF1A epitopes

    • Conduct antibody pre-absorption with recombinant TNFRSF1A before staining

  • Functional correlation analysis:

    • Correlate TNFRSF1A detection with downstream functional readouts (e.g., NF-κB activation)

    • Compare antibody binding with TNF-α-induced biological effects

    • Assess interference patterns with soluble TNFRSF1A versus membrane-bound forms

  • Cross-platform validation strategy:

    • Compare results between flow cytometry and immunohistochemistry

    • Correlate FITC signal with other detection methods (e.g., unconjugated primary with secondary detection)

    • Validate findings with orthogonal methods like mass cytometry or spectral flow cytometry

  • Advanced multiplexing controls:

    • Perform multi-color staining with antibodies to known TNFRSF1A-associated proteins

    • Conduct phospho-flow analysis correlating receptor expression with activation of downstream pathways

    • Implement co-localization studies with TNF-α ligand binding

This systematic approach provides multiple layers of evidence for antibody specificity and performance reliability across different experimental conditions.

What are common pitfalls in flow cytometry experiments using FITC-conjugated TNFRSF1A antibodies?

Flow cytometry with FITC-conjugated TNFRSF1A antibodies presents several technical challenges that researchers frequently encounter. Understanding these pitfalls and their solutions is critical for generating reliable data:

  • Signal-to-noise ratio problems:

    • Cause: Suboptimal antibody concentration or insufficient blocking

    • Solution: Perform systematic titration experiments (typically testing 0.008-0.04 μg/mL range) and optimize blocking protocols with 5-10% serum matching the host species

  • FITC spectral overlap issues:

    • Cause: Inadequate compensation with other fluorophores, particularly PE

    • Solution: Use single-stained controls with the specific FITC-conjugated TNFRSF1A antibody rather than generic compensation beads to account for the particular fluorophore:protein ratio

  • Receptor modulation during processing:

    • Cause: TNFRSF1A shedding or internalization during sample preparation

    • Solution: Minimize processing time, maintain samples at 4°C, and consider adding metalloproteinase inhibitors to prevent receptor shedding

  • Fixation-induced epitope masking:

    • Cause: Some fixatives can alter TNFRSF1A epitope conformation

    • Solution: Compare multiple fixation protocols (paraformaldehyde concentrations between 0.5-2%) or consider using live-cell staining protocols when possible

  • Clone-specific binding limitations:

    • Cause: Different antibody clones (e.g., H398 vs. 16803 ) recognize distinct epitopes

    • Solution: Test multiple antibody clones when establishing new cell systems or examining TNFRSF1A in different conformational states

  • Sample-specific autofluorescence:

    • Cause: Certain cell types (macrophages, dendritic cells) have high autofluorescence in the FITC channel

    • Solution: Implement autofluorescence reduction protocols (e.g., Trypan blue quenching) or switch to alternative fluorophores with emission spectra outside the autofluorescence range

  • Data analysis misinterpretation:

    • Cause: Inappropriate gating strategies based solely on isotype controls

    • Solution: Use fluorescence-minus-one (FMO) controls for more accurate gate placement and include biological controls (known positive and negative populations)

How can researchers effectively measure both membrane-bound and soluble forms of TNFRSF1A?

TNFRSF1A exists in both membrane-bound and soluble forms, with the latter resulting from proteolytic cleavage of the receptor ectodomain . Developing comprehensive experimental approaches to quantify both forms requires specific methodological considerations:

  • Membrane-bound TNFRSF1A detection:

    • Flow cytometry with FITC-conjugated antibodies targeting the extracellular domain

    • Cell surface biotinylation followed by immunoprecipitation and Western blotting

    • Immunofluorescence microscopy with careful membrane visualization techniques

  • Soluble TNFRSF1A quantification:

    • Custom sandwich ELISA using capture antibodies targeting different epitopes than detection antibodies

    • Bead-based multiplex assays allowing simultaneous measurement of soluble TNFRSF1A and related cytokines

    • Immunoprecipitation from conditioned media or biological fluids followed by Western blotting

  • Integrated dual-detection approaches:

    • Sequential isolation protocols separating membrane fractions from soluble fractions before analysis

    • Time-course experiments measuring receptor shedding rates by monitoring both forms simultaneously

    • Reporter cell lines expressing fluorescently-tagged TNFRSF1A for live monitoring of shedding kinetics

  • Ratio analysis methodology:

    • Calculate membrane-to-soluble ratio as a dynamic measure of receptor processing

    • Compare ratios across different experimental conditions and disease states

    • Correlate ratio shifts with functional outcomes in cellular response assays

  • Shedding modulation experiments:

    • Use metalloproteinase inhibitors (e.g., TAPI-0, TAPI-1) to block TNFRSF1A shedding

    • Implement stimulation protocols with PMA to enhance shedding

    • Compare natural shedding patterns with interventional approaches

  • Clinical sample considerations:

    • Optimize pre-analytical handling to preserve both receptor forms

    • Standardize collection protocols for longitudinal monitoring

    • Implement appropriate calibration standards for absolute quantification

This comprehensive approach enables researchers to understand the dynamic relationship between receptor forms and their functional significance in various biological contexts.

What strategies maximize the detection sensitivity of low-abundance TNFRSF1A in tissue samples?

Detecting low-abundance TNFRSF1A in tissue samples presents significant technical challenges requiring specialized approaches to enhance sensitivity while maintaining specificity:

  • Signal amplification technologies:

    • Tyramide signal amplification (TSA): Can increase FITC signal 10-100 fold by catalyzing local deposition of additional fluorophores

    • Polymer-based detection systems: Employ high-density fluorophore-conjugated polymers for enhanced signal

    • Sequential multi-layer detection: Apply biotinylated secondary antibodies followed by fluorescent streptavidin conjugates

  • Advanced sample preparation protocols:

    • Antigen retrieval optimization: Compare heat-induced (citrate, EDTA, Tris buffers at pH 6.0-9.0) and enzyme-based methods

    • Extended primary antibody incubation: Increase from standard 1-2 hours to overnight at 4°C

    • Signal-to-noise enhancement: Implement extended blocking (3-5% BSA, 10% serum) and multiple washing steps

  • Instrument optimization for FITC detection:

    • Confocal microscopy: Utilize spectral detectors with optimized bandpass filters matching FITC emission spectrum

    • Flow cytometry: Increase PMT voltage while maintaining negative population separation

    • Digital imaging: Employ computational deconvolution algorithms to enhance signal discrimination

  • Combined probe approaches:

    • Dual-antibody staining: Use multiple TNFRSF1A antibodies recognizing different epitopes

    • Combined RNA/protein detection: Implement RNAscope with immunofluorescence to correlate transcript and protein

    • Proximity ligation assay: Detect TNFRSF1A interaction with binding partners for functional verification

  • Sample enrichment methods:

    • Laser capture microdissection: Isolate regions with suspected TNFRSF1A expression

    • Cell sorting: Enrich for specific populations prior to analysis

    • Receptor internalization inhibition: Block endocytosis to maximize surface retention

  • Controls for validation:

    • Tissue-matched TNFRSF1A overexpression models

    • Concentration curves with recombinant protein spiking

    • Correlation with mass spectrometry-based proteomic quantification

These approaches can be systematically implemented and optimized according to specific tissue characteristics and research questions.

How does receptor internalization affect the detection and functional analysis of TNFRSF1A?

TNFRSF1A undergoes dynamic internalization following ligand binding, which significantly impacts both detection and functional studies. Understanding these dynamics is crucial for accurate experimental design and interpretation:

  • Internalization kinetics and mechanistic considerations:

    • TNFRSF1A typically internalizes within 5-30 minutes following TNF-α binding

    • Clathrin-dependent and lipid raft-mediated pathways both contribute to internalization

    • Internalized receptors may continue signaling from endosomal compartments

  • Detection challenges and solutions:

    • Surface vs. total detection: Compare non-permeabilized vs. permeabilized conditions to distinguish populations

    • Kinetic analysis: Implement time-course experiments with synchronized receptor stimulation

    • Compartment-specific detection: Use co-localization with endosomal markers (EEA1, Rab5, Rab7)

  • Inhibitor-based experimental approaches:

    • Pharmacological intervention: Apply dynamin inhibitors (Dynasore), clathrin inhibitors (Pitstop-2), or endosomal acidification inhibitors (Bafilomycin A1)

    • Temperature manipulation: Perform staining at 4°C to inhibit internalization processes

    • Genetic approaches: Express dominant-negative dynamin or Eps15 constructs

  • Functional consequence analysis:

    • Signaling compartmentalization: Compare NF-κB activation kinetics with and without internalization inhibitors

    • Temporal signaling patterns: Measure how internalization affects the duration of downstream signaling

    • Cell-type specific differences: Compare internalization rates and consequences across immune, epithelial and other cell types

  • Advanced microscopy approaches:

    • Live-cell imaging with pH-sensitive fluorophores to track receptor movement through acidifying compartments

    • Super-resolution microscopy to visualize receptor clustering prior to internalization

    • FRET-based sensors to detect conformational changes during trafficking

  • Correlative internalization-degradation analysis:

    • Pulse-chase experiments to track receptor fate after internalization

    • Lysosomal inhibitor studies to determine degradation contribution

    • Recycling rate quantification using surface biotinylation stripping assays

These methodologies provide a comprehensive framework for investigating how TNFRSF1A internalization dynamics influence both experimental detection and biological function interpretation.

How can FITC-conjugated TNFRSF1A antibodies be effectively utilized in multiplexed flow cytometry panels?

Incorporating FITC-conjugated TNFRSF1A antibodies into multiplexed flow cytometry panels requires careful consideration of spectral compatibility, panel design, and optimization strategies:

  • Spectral compatibility planning:

    • FITC emission profile: Peak at ~520nm with significant overlap with PE (~575nm)

    • Recommended fluorochrome combinations: Pair with APC, APC-Cy7, PE-Cy7 and BV421 to minimize compensation requirements

    • Avoid or carefully compensate: PE, PerCP, and BB515 due to spectral overlap

  • Panel design strategies for different research questions:

    • Immunophenotyping: Combine FITC-TNFRSF1A with lineage markers on separate channels (CD3-APC, CD19-PE-Cy7, CD14-BV421)

    • Signaling pathway analysis: Include phospho-specific antibodies (p-NF-κB p65-PE-Cy7, p-p38 MAPK-APC)

    • Receptor family analysis: Examine multiple TNF-receptors simultaneously (TNFRSF1A-FITC, TNFRSF1B-APC, FAS-BV421)

  • Titer optimization in multiplexed context:

    • Perform sequential titration: First optimize FITC-TNFRSF1A alone, then in presence of other antibodies

    • Evaluate spillover spreading error: Test antibody performance at multiple PMT voltages

    • Adjust concentration based on competition: May require higher concentrations in full panels than when used individually

  • Advanced compensation strategies:

    • Single-stained controls: Use cells rather than beads when possible, matching the biological sample type

    • Automated compensation algorithms: Apply software-based compensation with manual verification

    • Compensation stability: Validate compensation matrix stability across multiple experimental days

  • Panel-specific sample preparation considerations:

    • Buffer optimization: Test commercial buffers specifically designed for multicolor flow cytometry

    • Fixation timing: Determine optimal fixation duration that preserves all epitopes in the panel

    • Sequential staining: Consider step-wise protocols for markers with potential interference

  • Analytical approaches for complex panels:

    • Dimensionality reduction: Apply tSNE or UMAP for visualization of high-parameter data

    • Automated clustering: Use FlowSOM or similar algorithms to identify populations objectively

    • Correlation analysis: Examine relationships between TNFRSF1A and other markers across identified clusters

This systematic approach ensures optimal performance of FITC-conjugated TNFRSF1A antibodies within complex multiparameter flow cytometry panels.

What are the key considerations for using TNFRSF1A antibodies in immunohistochemistry of different tissue types?

Applying TNFRSF1A antibodies to diverse tissue types for immunohistochemistry requires tissue-specific adaptations to optimize detection while minimizing artifacts:

  • Tissue-specific fixation and processing protocols:

    • Lymphoid tissues (spleen, lymph nodes): Shorter fixation times (6-12 hours in 10% neutral buffered formalin)

    • Kidney samples: Extended fixation (12-24 hours) with careful pH monitoring

    • Adipose tissue: Special fixatives with reduced alcohol content to preserve membrane structures

    • Brain tissue: Post-fixation cryoprotection to maintain antigenicity while preserving structure

  • Antigen retrieval optimization for each tissue type:

    • Kidney: Heat-induced epitope retrieval in Tris-EDTA buffer (pH 9.0) often yields optimal results

    • Lymphoid tissue: Enzyme-based retrieval (proteinase K) may better expose TNFRSF1A epitopes

    • Lung tissue: Citrate buffer (pH 6.0) with controlled heating rates to prevent tissue destruction

    • Liver: Combined approaches with both heat and enzymatic treatment for difficult samples

  • Detection system selection based on tissue characteristics:

    • High autofluorescence tissues (kidney, brain): Consider chromogenic detection systems or fluorophores outside the FITC spectrum

    • Low expression tissues: Multi-step detection with biotin-streptavidin amplification

    • Tissues with high endogenous biotin (liver, kidney): Implement avidin-biotin blocking steps

  • Tissue-specific background reduction strategies:

    • Kidney: Extended peroxidase blocking (3% H2O2, 15-30 minutes) and additional serum blocking

    • Adipose tissue: Include lipid removal steps prior to antibody incubation

    • Brain: Apply Sudan Black B treatment to reduce lipofuscin autofluorescence

    • Skin: Extended blocking with normal serum matching secondary antibody species

  • Validation controls tailored to tissue context:

    • Tissue-specific positive controls: Use samples with known TNFRSF1A expression patterns (e.g., kidney glomeruli and tubulointerstitium)

    • Absorption controls: Pre-incubate antibodies with recombinant TNFRSF1A before tissue application

    • Competing structures control: Test for non-specific binding to tissue-specific structural elements

  • Specialized counterstaining approaches:

    • Nuclear counterstains: Adjust concentration and incubation time based on tissue density

    • Multi-color co-localization: Combine with cell-type-specific markers optimal for each tissue

    • Basement membrane visualization: Add special stains to delineate structural boundaries relevant to TNFRSF1A localization

These tissue-specific adaptations maximize the reliability of TNFRSF1A detection across diverse histological specimens.

How can TNFRSF1A antibodies contribute to studying receptor signaling dynamics in different disease contexts?

TNFRSF1A antibodies provide powerful tools for investigating the complex signaling dynamics of this receptor across various pathological conditions:

  • Temporal signaling analysis in inflammatory diseases:

    • Kinetic profiling: Track TNFRSF1A expression, internalization, and degradation rates during disease progression

    • Signaling cascade visualization: Combine with phospho-specific antibodies to map activation sequence

    • Receptor cross-talk: Examine how TNFRSF1A signaling interacts with other inflammatory pathways in a disease-specific manner

  • Cancer research applications:

    • Tumor microenvironment: Compare TNFRSF1A signaling in tumor cells versus infiltrating immune cells

    • Therapy response prediction: Correlate TNFRSF1A expression patterns with response to immunotherapies

    • Resistance mechanism investigation: Examine alterations in receptor expression, localization, and downstream signaling in treatment-resistant tumors

  • Autoimmune disease investigation:

    • Cell-specific profiling: Quantify TNFRSF1A on different immune cell subsets in patients versus controls

    • Therapy monitoring: Track changes in receptor dynamics during anti-TNF treatment

    • Biomarker development: Correlate soluble versus membrane-bound TNFRSF1A ratios with disease activity

  • Cardiovascular pathology research:

    • Endothelial activation: Measure TNFRSF1A-triggered signaling in vascular endothelium during atherosclerosis

    • Cardiac remodeling: Investigate receptor signaling in cardiomyocytes during heart failure progression

    • Therapeutic intervention assessment: Evaluate how modulating TNFRSF1A affects cardiovascular outcomes

  • Neurodegenerative disease applications:

    • Neuroinflammation monitoring: Track microglial TNFRSF1A activation patterns in disease models

    • Blood-brain barrier studies: Examine how receptor signaling affects barrier permeability

    • Neuroprotection strategies: Test how selective TNFRSF1A signaling modulation affects neuronal survival

  • Advanced single-cell approaches:

    • Mass cytometry integration: Incorporate TNFRSF1A detection into CyTOF panels for high-dimensional phenotyping

    • Single-cell sequencing correlation: Link receptor protein expression with transcriptional signatures

    • Spatial proteomics: Map receptor distribution within tissue microenvironments using multiplexed imaging

These applications highlight how TNFRSF1A antibodies can address fundamental questions about receptor biology in disease pathogenesis and treatment response.

What emerging technologies can enhance the utility of FITC-conjugated TNFRSF1A antibodies in research?

Several cutting-edge technologies are expanding the capabilities and applications of FITC-conjugated TNFRSF1A antibodies in immunological research:

  • Advanced microscopy platforms:

    • Lattice light-sheet microscopy: Enables long-term, high-resolution imaging of TNFRSF1A dynamics in living cells with minimal phototoxicity

    • Super-resolution techniques (STORM, PALM): Achieve 10-20nm resolution to visualize receptor nanoclusters and molecular organization

    • Expansion microscopy: Physical sample expansion allows standard confocal microscopes to achieve super-resolution imaging of TNFRSF1A distribution

  • Enhanced flow cytometry approaches:

    • Spectral flow cytometry: Better separation of FITC from autofluorescence and other fluorophores through full spectral analysis

    • Imaging flow cytometry: Combines spatial information with quantitative analysis for subcellular localization of TNFRSF1A

    • High-throughput flow cytometry: Automated sampling for screening TNFRSF1A expression across large cell libraries or patient cohorts

  • Microfluidic and organ-on-chip systems:

    • Single-cell secretion analysis: Correlate TNFRSF1A expression with cytokine production at individual cell level

    • Biomimetic tissue interfaces: Study TNFRSF1A signaling in complex multicellular environments

    • Gradient generation platforms: Examine receptor response to controlled TNF-α concentration gradients

  • Nanoparticle-based technologies:

    • Antibody-conjugated quantum dots: Provide enhanced brightness and photostability for long-term tracking

    • SERS-active nanoparticles: Label antibodies for multiplexed detection beyond fluorescence limitations

    • DNA-barcoded antibodies: Enable ultrahigh-multiplexing for simultaneous detection of TNFRSF1A and hundreds of other targets

  • Artificial intelligence integration:

    • Automated image analysis: Implement deep learning for objective quantification of receptor patterns

    • Predictive modeling: Correlate imaging features with functional outcomes

    • Multiparametric data integration: Combine antibody-generated data with other -omics approaches

  • Novel bioconjugation strategies:

    • Site-specific conjugation: Target specific antibody residues to maintain optimal antigen binding

    • Cleavable linkers: Design antibody-fluorophore connections responsive to specific cellular environments

    • Photoactivatable fluorophores: Enable precise spatiotemporal control of FITC visualization

These technological advances are dramatically expanding the research applications and analytical depth possible with FITC-conjugated TNFRSF1A antibodies.

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.