Recombinant Human Tumor necrosis factor protein (TNF), partial (Active)

Shipped with Ice Packs
In Stock

Description

Biological Activity and Mechanism

The partial TNF retains pro-inflammatory and cytotoxic functions:

  • Apoptosis induction: Activates TNFR1-mediated apoptosis in cancer cells (e.g., Du145 cells) via caspase activation .

  • Trimer structure: Essential for high-affinity receptor binding (KD = 90 pM for trimers vs. 70 nM for monomers) .

  • Selective targeting: Engineered variants like RMP16 (TNF-α-derived peptide with albumin-binding domain) exhibit prolonged half-life (13.1 h) and enhanced tumor specificity .

Key Receptors and Functions

ReceptorFunctionReference
TNFR1Apoptosis, inflammation, and immune cell activation (ubiquitous expression)
TNFR2Proliferation, survival, and vascular effects (hematopoietic cells)

Cancer Therapy

  • Tumor necrosis: Direct cytotoxicity against malignant cells (e.g., breast, colorectal, lung cancer) .

  • Synergy with chemotherapy: Enhances drug delivery via vascular permeability (EPR effect) .

  • RMP16 variant: Inhibits angiogenesis and tumor growth by selectively activating TNFR1 .

Immunological Studies

  • Inflammation models: Induces fever, cachexia, and cytokine cascades (e.g., IL-1, IFN-γ) .

  • Reporter cell systems: Validated using HEK-Blue™ TNF-α cells for inhibitor screening .

Expression Systems

HostAdvantagesLimitations
E. coliHigh yield, cost-effectiveLack of glycosylation; inclusion bodies
YeastProper folding; post-translational modifications (e.g., disulfide bonds)Lower yield; complex purification
Mammalian (CHO)Native glycosylation; improved bioactivityHigh production costs

Purification and Validation

  • Affinity chromatography: Utilizes His-tag for nickel or cobalt resin binding .

  • Bioactivity assays: Confirmed via cytotoxicity (ED₅₀) and receptor binding (SPR/ITC) .

Pharmacological Advances

  • Half-life extension: RMP16’s albumin-binding domain increases circulation time (13.1 h vs. TNF-α’s 29 min) .

  • Anti-TNF therapies: Monoclonal antibodies (e.g., adalimumab) neutralize TNF in autoimmune diseases .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 20 mM PB, 10 mM NaCl, pH 7.0
Description

Recombinant human tumor necrosis factor protein (TNF) is a partial-length protein comprising amino acids 77-233 of human TNF. Expressed in E. coli cells, this protein exhibits a purity exceeding 98%, as confirmed by SDS-PAGE and HPLC analysis. Functional characterization through a cytotoxicity assay using murine L929 cells demonstrates its full biological activity (ED50 <0.05 ng/ml, specific activity >2.0x107 IU/mg). Endotoxin levels are below 1.0 EU/µg, as determined by the LAL method. Our inventory includes ready-to-use TNF proteins, and custom tag modifications are available upon request. This recombinant TNF protein serves as a valuable tool for various applications, including specific antibody synthesis and cancer research.

TNF, also known as TNF-α, is a pro-inflammatory cytokine that plays a crucial role in the inflammatory response. Its pro-inflammatory effects are mediated via TNF-α/TNFR signaling. TNF is primarily produced by macrophages and is triggered in response to injury, bacterial or viral infections. Its function is to protect against infections and promote wound healing. However, excessive TNF expression can lead to excessive inflammation and may contribute to the development of autoimmune diseases. Several inflammatory autoimmune conditions, including psoriatic arthritis, rheumatoid arthritis (RA), and Crohn's disease, are linked to TNF. Anti-TNF therapy has become a standard clinical treatment for managing autoimmune diseases like RA. Furthermore, research has demonstrated that TNF can induce cell death in certain tumor cells.

Form
Liquid or Lyophilized powder
Lead Time
5-10 business days
Shelf Life
The shelf life of this product is influenced by several factors, including storage conditions, buffer composition, temperature, and the protein's intrinsic stability. Generally, liquid forms maintain stability for 6 months at -20°C/-80°C, while lyophilized forms exhibit a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. For multiple use, aliquoting is recommended to minimize freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
APC1; APC1 protein; Cachectin; DIF; Differentiation inducing factor; Macrophage cytotoxic factor; Tnf; TNF superfamily member 2; TNF superfamily, member 2; TNF, macrophage derived; TNF, monocyte derived; TNF-a; TNF-alpha; TNFA; TNFA_HUMAN; TNFSF2; Tumor necrosis factor (TNF superfamily member 2); Tumor necrosis factor alpha; Tumor necrosis factor; Tumor necrosis factor ligand superfamily member 2; Tumor Necrosis Factor, Membrane Form; Tumor necrosis factor, soluble form
Datasheet & Coa
Please contact us to get it.
Expression Region
77-233aa
Mol. Weight
17.5 kDa
Protein Length
Partial
Purity
>98% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
TNF
Uniprot No.

Target Background

Function

Tumor necrosis factor (TNF) is a cytokine that binds to TNFRSF1A/TNFR1 and TNFRSF1B/TNFBR. It is primarily secreted by macrophages and can induce cell death in certain tumor cell lines. TNF is a potent pyrogen, causing fever directly or by stimulating interleukin-1 secretion. Additionally, it is implicated in the development of cachexia. Under specific conditions, TNF can promote cell proliferation and induce cell differentiation. TNF impairs the function of regulatory T-cells (Treg) in individuals with rheumatoid arthritis by dephosphorylating FOXP3, a key transcription factor in Treg cells. This dephosphorylation inactivates FOXP3, rendering the Treg cells functionally defective. TNF upregulates the expression of protein phosphatase 1 (PP1), which is responsible for dephosphorylating FOXP3. TNF is a key mediator of cell death in the anticancer action of BCG-stimulated neutrophils in combination with DIABLO/SMAC mimetic in the RT4v6 bladder cancer cell line. TNF induces insulin resistance in adipocytes by inhibiting insulin-induced IRS1 tyrosine phosphorylation and insulin-induced glucose uptake. It also induces GKAP42 protein degradation in adipocytes, contributing to TNF-induced insulin resistance. TNF plays a role in angiogenesis by inducing VEGF production synergistically with IL1B and IL6. The intracellular domain (ICD) form of TNF induces IL12 production in dendritic cells.

Gene References Into Functions
  1. Genetic predisposition to rheumatoid arthritis in the Russian population in the Republic of Karelia is associated with the presence of the GG TNF-alpha genotype. PMID: 30225702
  2. Treatment with 30 microg/ml curcumin significantly reduces TNFalpha protein production in Behcet's disease patients (p < .01) and healthy controls (p < .05) M1 macrophages. PMID: 29806793
  3. The A allele in TNF-alpha 238A/G, but not TNF-alpha 308A/G, demonstrates a protective role against the occurrence of juvenile idiopathic arthritis in the Caucasian population (Meta-Analysis). PMID: 30412082
  4. TNF-alpha expressed regionally in Epicardial Adipose Tissue may exert potent effects on the progression of coronary atherosclerosis. PMID: 28931782
  5. The effect of a pro-inflammatory diet on TNF-alpha concentrations was more pronounced in pregnant women reporting higher levels of stress. PMID: 30200631
  6. TNF-alpha -308G/A polymorphism is not associated with the risk of developing bullous pemphigoid and alopecia areata in an Iranian cohort. PMID: 29843231
  7. Data suggest that only the promoter single-nucleotide polymorphism (SNP) rs1800629 within the TNF-alpha gene is associated with an increased risk for developing Graves' disease (GD), especially in the European population. Larger-scale studies are required to validate the associations between TNF-alpha gene and GD. PMID: 29440561
  8. A study involving 173 polymorphisms investigated the association with the response to anti-TNF drugs in patients with moderate-to-severe plaque psoriasis (N=144). PMID: 27670765
  9. The SNP rs4819554 in the promoter region of IL17RA significantly influences the response to anti-TNF drugs at week 12. PMID: 27670766
  10. TNF-alpha promoter gene polymorphisms and/or haplotypes are risk factors for Nephrotic syndrome and resistance to steroids among Egyptian children. PMID: 28803697
  11. The findings suggest that polymorphism in the TNF-alpha gene might be a risk factor for nasal polyposis in the northern part of Iran, with a minor frequency of the G308A allele slightly higher than in other major populations. PMID: 30003390
  12. Results indicate that the interplay of pro-inflammatory cytokines IFN-gamma derived from CD4+T lymphocytes and TNF-alpha from CD14+ cells does not directly contribute to parasite replication but induces IL-4 production. PMID: 29953494
  13. Single nucleotide polymorphisms rs361525, rs1800629, and rs17999645 of tumor necrosis factor-alpha were significantly correlated with the diagnosis of cervical cancer. PMID: 29940817
  14. Rs11541076 in IRAK3, a negative regulator of TLR signaling, is a predictor of anti-TNF treatment response. PMID: 27698401
  15. In this Brazilian population, TNF and IL17 gene polymorphisms responsible for the expression of important inflammatory cytokines were associated with overall spondyloarthritis, specifically ankylosing spondylitis and psoriatic arthritis, regardless of gender and HLA-B27. PMID: 29849482
  16. By restraining TNFR1 at the cell surface via sialylation, ST6Gal-I acts as a functional switch to divert signaling toward survival. These findings point to a novel glycosylation-dependent mechanism that regulates the cellular response to TNF and may promote cancer cell survival within TNF-rich tumor microenvironments. PMID: 29233887
  17. The TNF*A allele confers susceptibility to AIH in Tunisian patients and is associated with increased production of TNF-alpha. Anti-TNF antibodies could be an alternative to corticotherapy and may avoid the exacerbated immune response in Autoimmune hepatitis. PMID: 29845365
  18. Impairment in TNF, IL-1beta, and IL-17 production upon stimulation with mycobacterial antigens may contribute to the increased susceptibility to M. tuberculosis infection observed in HTLV-1 infected individuals. PMID: 29523325
  19. LL was significantly negatively correlated with PGC-1alpha, TNF-alpha, and IL-6 mRNA expressions. PGC-1alpha mRNA expression levels in paraspinal muscles may be affected by lumbar kyphosis. PMID: 30233161
  20. TNF-alpha-308G>A polymorphism affects the overall survival of cancer patients and is a potential therapeutic target for cancer. PMID: 30407345
  21. Many inflammatory pathologies are now recognized to be driven by aberrant TNF-induced cell death, which, in most circumstances, depends on the kinase Receptor-interacting serine/threonine-protein kinase 1 (RIPK1). [review] PMID: 29217118
  22. High TNF-alpha expression is associated with retinopathy of prematurity. PMID: 29274846
  23. Tumor necrosis factor-alpha selectively reduces BMPR-II transcription and mediates post-translational BMPR-II cleavage via the sheddases, ADAM10 and ADAM17 in pulmonary artery smooth muscle cells. PMID: 28084316
  24. Polymorphisms of IL-1betab and TNF-a are not a risk of ICC, but an individual with O. viverrini infection has an effect on all genotypes of the TNF-alpha gene that might promote intrahepatic cholangiocarcinoma. Primary prevention of intrahepatic cholangiocarcinoma in high-risk areas is based on efforts to reduce O. viverrini infection. PMID: 30139338
  25. In the placenta, when gestational age was controlled for, partial correlation revealed a significant positive correlation between TNF-alpha and MMP-9 only in the second trimester. PMID: 28820024
  26. Study shows that in human endometrial stromal cells, high tumor necrosis factor levels negatively affect the insulin action through decreased adiponectin signaling and glucose transporter type 4 protein. This could explain the failures observed in endometrial function of obese women with polycystic ovary syndrome. PMID: 28946816
  27. Three single nucleotide polymorphisms (SNPs) within P2X4R and two SNPs within CAMKK2 influenced concentrations of TNFalpha in peripheral blood mononuclear cells, but these SNP did not associate with the risk for HIV-associated sensory neuropathy in South Africans. PMID: 29428485
  28. Results indicated that the proinflammatory cytokine TNF-alpha impairs endothelial tight junctions and promotes monocyte-endothelial cell adhesion by upregulating beta-site amyloid precursor protein enzyme 1 expression through activating PKC signaling and sequentially cleaving alpha-2, 6-sialic acid transferase 1. PMID: 28091531
  29. These findings reveal that PrP enhances the responses to TNF-alpha, promoting proinflammatory cytokine production, which may contribute to inflammation and tumorigenesis. PMID: 28900035
  30. This study found that the protein and mRNA expression levels of the cytokines TNF-alpha are significantly increased. PMID: 28476335
  31. Taking together, these results suggest that Wnt/beta-catenin signal pathway activation-dependent up-regulation of syncytin-1 contributes to the pro-inflammatory factor TNF-alpha-enhanced fusion between oral squamous cell carcinoma cells and endothelial cells. PMID: 28112190
  32. Data suggest that, in children with pediatric obesity, lifestyle weight-loss intervention results in down-regulation of serum cardiotrophin-1 (CTF1), interleukin-6 (IL6), and tumor necrosis factor-alpha (TNFA); expression of CTF1, IL6, and TNFA is also down-regulated in peripheral blood mononuclear cells after improvement in adiposity, body mass index, and waist-hip ratio. PMID: 28749076
  33. TNFalpha differently regulated the levels of PPARalpha, LXRalpha, and LXRbeta binding to the apoA-I gene promoter in THP-1 cells. Obtained results suggest a novel tissue-specific mechanism of the TNFalpha-mediated regulation of apoA-I gene in monocytes and macrophages and show that endogenous ApoA-I might be positively regulated in macrophage during inflammation. PMID: 29442267
  34. This is the first evidence to suggest that TET2 mutations promote clonal dominance with aging by conferring TNFalpha resistance to sensitive bone marrow progenitors while also propagating such an inflammatory environment. PMID: 29195897
  35. Anti-rotavirus effect of TNF-alpha was achieved by NFkappaB-regulated genes via the activation of classical nuclear factor kappaB (NF-kappaB) signaling. PMID: 29859235
  36. Results revealed that heterozygous genotype GA of TNF-alpha-238 (rs 361525) SNP significantly increased the risk of adverse-outcome (mortality rate), regardless of organ dysfunction or severity of sepsis. PMID: 29978383
  37. In patients with primary depression, depressive symptoms were associated with TNF-alpha. PMID: 30148175
  38. Addition of TNFalpha to podocytes causes CD80 upregulation, actin reorganization, and podocyte injury. PMID: 29022109
  39. The results of the study suggest that the levels of C-reactive protein and tumor necrosis factor-alpha are important diagnostic markers of inflammation in patients with chronic pancreatitis and type 2 diabetes mellitus. PMID: 30280549
  40. TNF-alpha and IL-10 treatment can affect the expression of ICAM-1 and CD31 in human coronary artery endothelial cells. PMID: 29949812
  41. The present study demonstrated the ability of 30 and 100 ng/ml TIMP3 to attenuate migration and proliferation, and to inhibit the activity of MMP2, MMP9 and TNFalpha secretion of NA SMCs. In conclusion, TIMP3 may be considered a potential therapeutic drug for use in a novel drugeluting stent, to attenuate the progressive dilation of the aortic NA. PMID: 29956789
  42. Elevated A20 promotes TNF-induced and RIPK1-dependent intestinal epithelial cell death. PMID: 30209212
  43. TNF-alpha GG genotype at -238 and GG haplotype at positions -308 and -238 were associated with Kawasaki disease in an Iranian population. PMID: 27455075
  44. We have shown that a TNFalpha gene polymorphism, rs1800629, is highly significantly associated with postmenopausal osteoporosis and BMD in the female Han Chinese population. PMID: 29481288
  45. Allele -308 A TNF-alpha may have a role in the progression of rheumatoid arthritis in a North Indian population. PMID: 28748515
  46. ATF3 mediates the inhibitory action of TNF-alpha on osteoblast differentiation, and the TNF-alpha-activated JNK pathway is responsible for the induction of Atf3 expression. PMID: 29605296
  47. TNF-alpha, DKK1, and OPG have roles in the pathogenesis of knee osteoarthritis. PMID: 28676900
  48. Btk acts in the TLR7/8 pathway and mediates Ser-536 phosphorylation of p65 RelA and subsequent nuclear entry in primary human macrophages. PMID: 29567473
  49. Results indicated that prolonged tumor necrosis factor (TNFalpha) exposure could have detrimental consequences to endothelial cells by causing senescence, therefore, chronically increased TNFalpha levels might possibly contribute to the pathology of chronic inflammatory diseases by driving premature endothelial senescence. PMID: 28045034

Show More

Hide All

Database Links

HGNC: 11892

OMIM: 191160

KEGG: hsa:7124

STRING: 9606.ENSP00000398698

UniGene: Hs.241570

Involvement In Disease
Psoriatic arthritis (PSORAS)
Protein Families
Tumor necrosis factor family
Subcellular Location
Cell membrane; Single-pass type II membrane protein.; [Tumor necrosis factor, membrane form]: Membrane; Single-pass type II membrane protein.; [Tumor necrosis factor, soluble form]: Secreted.; [C-domain 1]: Secreted.; [C-domain 2]: Secreted.

Q&A

What is the molecular structure of recombinant human TNF-alpha?

Recombinant human TNF-alpha is typically expressed as an E. coli-derived protein spanning Val77-Leu233 of the native sequence, with or without an N-terminal Met residue. The functional protein exists as a homotrimer with a molecular weight of approximately 53.1 kDa as determined by SEC-MALS analysis, although the monomeric form appears as a 17 kDa band under reducing SDS-PAGE conditions . The complete native human TNF-alpha consists of a 35 amino acid cytoplasmic domain, a 21 amino acid transmembrane segment, and a 177 amino acid extracellular domain (ECD) . For research purposes, it's critical to note that the recombinant form represents the soluble, cleaved portion of the protein that corresponds to the bioactive extracellular domain.

How does recombinant human TNF-alpha compare structurally with TNF-alpha from other species?

Within the extracellular domain, human TNF-alpha shares 97% amino acid sequence identity with rhesus macaque TNF-alpha and between 71-92% identity with TNF-alpha from bovine, canine, cotton rat, equine, feline, mouse, porcine, and rat sources . This high degree of conservation reflects the evolutionary importance of this cytokine. When designing cross-species experiments, researchers should account for these homology differences, particularly when evaluating receptor binding specificity or developing neutralizing antibodies.

What is the standard method for measuring TNF-alpha bioactivity?

The standard bioassay for TNF-alpha activity utilizes the L-929 mouse fibroblast cytotoxicity assay in the presence of actinomycin D (a metabolic inhibitor). The effective dose (ED50) typically ranges from 25-100 pg/mL . This assay measures the cytotoxic potential of TNF-alpha and serves as the gold standard for activity determination. Alternative methods include receptor binding assays, reporter gene assays using NF-κB responsive elements, and measurement of specific downstream signaling events such as phosphorylation of IκB.

What are the optimal storage and handling conditions for maintaining recombinant TNF-alpha stability?

Lyophilized recombinant TNF-alpha should be stored at -20°C to -80°C. After reconstitution in sterile PBS containing at least 0.1% carrier protein (such as BSA), the protein remains stable for up to one month at 2-8°C or can be aliquoted and stored at -20°C to -80°C for up to 3 months with minimal loss of activity. Repeated freeze-thaw cycles should be avoided as they can lead to protein denaturation and activity loss. Working solutions should be prepared fresh on the day of experimentation whenever possible, and sterile technique should be employed to prevent microbial contamination.

How should TNF-alpha dose-response experiments be designed for cell-based assays?

When designing dose-response experiments with TNF-alpha, researchers should:

  • Establish a concentration range spanning at least 5 orders of magnitude (e.g., 0.1 pg/mL to 10 ng/mL)

  • Include a positive control (e.g., validated active TNF-alpha)

  • Determine cell type-specific responses, as sensitivity varies widely between cell types

  • Consider temporal dynamics, with typical response times ranging from 4-24 hours for transcriptional changes

  • Account for potential synergistic effects with other cytokines or stimuli

For cytotoxicity assays, the L-929 cell line with actinomycin D demonstrates optimal sensitivity with an ED50 of 25-100 pg/mL . For NF-κB activation assays in 293T cells, dose-response curves typically show activity in the nanomolar range, with compounds like S10 and 4e demonstrating IC50 values of 14 μM and 3 μM, respectively, when inhibiting TNF-induced activity .

What methodological approaches can be used to study TNF-alpha receptor binding?

Researchers can employ several complementary techniques to study TNF-alpha receptor binding:

  • Surface Plasmon Resonance (SPR): Allows real-time measurement of binding kinetics between immobilized TNFR and flowing TNF-alpha. This technique can detect competitive binding, as demonstrated in studies of small molecule inhibitors that block TNF-α and TNFR interactions .

  • Fluorescence Resonance Energy Transfer (FRET): Enables visualization of receptor-ligand interactions in living cells.

  • Radioligand binding assays: Provides quantitative measurement of binding affinity and receptor density.

  • Molecular modeling and docking: The crystal structure of TNF-α dimer (PDB code: 2AZ5) can be used for in silico prediction of binding interactions .

  • Cross-linking studies: Identifies specific binding sites and interaction domains.

Each method offers distinct advantages, and combining multiple approaches provides more robust characterization of receptor-ligand interactions.

How can recombinant TNF-alpha be used to investigate endothelial activation and procoagulant activity?

Recombinant TNF-alpha directly activates human vascular endothelial cells to induce tissue factor-like procoagulant activity (PCA). Methodologically, this can be studied through:

  • Isolation of primary endothelial cells from umbilical veins, saphenous veins, iliac arteries, or thoracic aortae

  • Treatment with rTNF at concentrations ranging from 100-500 units/ml

  • Measurement of PCA using one-stage clotting assays after 4-hour incubation

  • Assessment of PCA expression on intact viable endothelial monolayers

The response kinetics typically show a rapid rise to peak activity at approximately 4 hours, followed by a decline toward basal levels by 24 hours . This experimental system offers insights into thrombotic complications associated with inflammatory conditions. Importantly, the effects of TNF can be distinguished from those of IL-1 using specific neutralizing antisera, though their effects may be additive even at apparent maximal doses .

What are the pharmacokinetic parameters of recombinant TNF-alpha in vivo?

Clinical pharmacology studies with recombinant human TNF in cancer patients have established the following pharmacokinetic parameters:

ParameterValueNotes
Half-life20 minutesRapid clearance following IV administration
Initial serum concentration2.5 ng/mLAfter 35 μg/m² IV dose
Initial serum concentration80 ng/mLAfter 200 μg/m² IV dose
Therapeutic dose range (IV)1-200 μg/m²Used in clinical studies
Therapeutic dose range (SC)5-250 μg/m²Upper limit constrained by injection site inflammation

The rapid clearance suggests that continuous infusion or frequent dosing may be necessary to maintain therapeutic levels in experimental models . For research design, these parameters should inform dosing schedules and sampling timepoints, particularly in pharmacodynamic studies.

What are the most effective approaches for developing small molecule inhibitors targeting TNF-alpha?

Development of small molecule TNF-alpha inhibitors can follow this methodological framework:

  • Structure-based design utilizing the crystal structure of TNF-α dimer (PDB code: 2AZ5)

  • Shape screening of compound libraries using established inhibitors as templates

  • Rational design of derivatives based on structure-activity relationships

  • Combined in silico and experimental validation through:

    • SPR competitive binding assays between TNF-α and TNFR

    • Cell-based assays measuring NF-κB activation

    • Analysis of structural interactions using molecular docking

This approach has yielded compounds like 4e, a 2-oxo-N-phenyl-1,2-dihydrobenzo[cd]indole-6-sulfonamide derivative with an IC50 of 3.0 ± 0.8 μM in cell-based assays, representing one of the most potent TNF-α small molecule inhibitors reported . Researchers should focus on optimizing both binding affinity and pharmacokinetic properties when developing novel inhibitors.

What adverse effects should researchers consider when designing TNF-alpha experiments?

When designing experiments involving TNF-alpha administration, researchers should anticipate potential adverse effects based on clinical observations:

  • Constitutional symptoms: fever, chills, headache, fatigue

  • Cardiovascular effects: hypotension requiring fluid administration

  • Hematological changes: acute alterations in granulocyte, lymphocyte, and monocyte counts

  • Metabolic responses: changes in serum zinc levels and plasma cortisol consistent with acute phase response

  • Local inflammation: severe reactions at subcutaneous injection sites, particularly at doses of 125-250 μg/m²

  • Hemostatic alterations: minor changes in parameters related to coagulation

These effects should inform experimental design, particularly for in vivo studies, where appropriate monitoring and supportive measures should be planned . Researchers should implement the principle of refinement in animal studies by incorporating these considerations into humane endpoints and monitoring protocols.

How can researchers analyze TNF-alpha inhibitor safety profiles using pharmacovigilance data?

Analysis of TNF-alpha inhibitor safety can be approached using disproportionality analysis of pharmacovigilance databases such as the FDA Adverse Event Reporting System. The recommended statistical methods include:

  • Reporting Odds Ratio (ROR) with 95% confidence intervals

  • Information Component (IC) using Bayesian Confidence Propagation Neural Network

Signal detection criteria:

  • ROR025 (lower limit of 95% CI) > 1 and adverse event count > 3

  • IC025 (lower limit of 95% credible interval) > 0

Sensitivity analyses should include sex stratification to evaluate gender-specific signals . Visualization techniques such as UpSet plots can help identify common adverse events across different TNF inhibitors versus those unique to specific agents. This methodological approach provides systematic evidence for safety signal detection beyond anecdotal reports.

How can researchers address TNF-alpha experimental variability across different cell types?

Cell type-specific responses to TNF-alpha represent a significant source of experimental variability. To address this:

  • Characterize baseline expression levels of TNF receptors (TNFR1 and TNFR2) in your cell system using flow cytometry or Western blotting

  • Determine the relative contributions of each receptor using selective blocking antibodies

  • Assess the presence of soluble TNF receptors in your culture system that might neutralize TNF activity

  • Establish cell type-specific dose-response curves rather than relying on published ED50 values

  • Consider potential synergistic or antagonistic effects with other cytokines present in your experimental system

For primary cells, passage number and donor variability should be documented and accounted for in experimental design. Standardization of culture conditions (serum concentration, cell density, substrate) is essential for reproducible TNF-alpha responses.

What are the critical considerations when combining TNF-alpha with other cytokines or therapeutic agents?

When designing combination experiments with TNF-alpha, researchers should:

  • Determine sequence-dependent effects: the order of administration can significantly affect outcomes

  • Establish appropriate dose ratios based on the relative potencies of each agent

  • Account for potential receptor cross-talk and shared downstream signaling pathways

  • Consider temporal aspects of signaling dynamics for each agent

  • Validate additive, synergistic, or antagonistic effects using appropriate mathematical models (e.g., Chou-Talalay method)

Clinical studies suggest that combinations of TNF-alpha with other cytokines and cytostatic agents may produce enhanced therapeutic effects compared to TNF-alpha monotherapy . This principle likely extends to experimental systems, necessitating careful experimental design to distinguish mechanistic interactions from simple additive effects.

What are the emerging approaches for studying TNF-alpha structure-function relationships?

Advanced structural biology techniques are expanding our understanding of TNF-alpha's molecular mechanism:

  • Cryo-electron microscopy to visualize TNF-receptor complexes in near-native states

  • Hydrogen-deuterium exchange mass spectrometry to map dynamic protein interactions

  • Single-molecule imaging to track TNF-receptor clustering and signaling complex formation

  • Structural proteomics to identify post-translational modifications affecting activity

  • Computational molecular dynamics simulations to model conformational changes during receptor engagement

These approaches can help resolve outstanding questions about how TNF-alpha trimers engage with receptor complexes and how this leads to differential signaling outcomes. The application of these techniques could inform the development of more selective TNF modulators with improved therapeutic profiles.

How can genetic and pharmacological perturbation be combined to study TNF-alpha signaling networks?

An integrated approach combining genetic and pharmacological tools provides powerful insights into TNF-alpha biology:

  • CRISPR-Cas9 knockout/knockin models of specific pathway components

  • Small molecule inhibitors with defined mechanisms targeting discrete pathway nodes

  • Phosphoproteomic analysis to map signaling cascades with temporal resolution

  • Single-cell transcriptomics to capture population heterogeneity in responses

  • Optogenetic or chemically-inducible systems for precise temporal control of TNF signaling

This multi-modal approach can help deconvolute the complex signaling networks downstream of TNF-alpha receptors and identify critical nodes that might serve as therapeutic targets. Importantly, this strategy can reveal compensatory mechanisms that may limit the efficacy of TNF inhibitors in certain contexts.

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.