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

Shipped with Ice Packs
In Stock

Description

Introduction

Recombinant Human Tumor Necrosis Factor Protein (TNF), partial (Active) is a bioengineered fragment of TNF-α, encompassing amino acids 77–233 of the full-length transmembrane precursor. Expressed in yeast with an N-terminal 6xHis-tag for purification, this partial-length protein retains critical extracellular domain (ECD) regions essential for receptor binding and cytotoxic activity . It serves as a research tool for studying TNF signaling, receptor interactions, and inflammatory pathways.

Bioactivity and Functional Characteristics

ParameterValue/DescriptionSource
Cytotoxicity (ED₅₀)33.32–47.38 pg/mL (L-929 mouse fibroblasts + actinomycin D)
Receptor BindingBinds TNF-R1 and TNF-R2 (affinity not quantified in this construct)

The partial TNF retains bioactivity comparable to full-length TNF-α, inducing cytotoxicity in L-929 cells. Its activity is validated via standardized cytotoxicity assays, though receptor-binding kinetics differ from soluble TNF-α trimer .

Key Uses

  1. ELISA Standard:

    • Serves as a positive control in TNF-α detection assays.

    • Example: Used in functional ELISA with TNFSF13B-Biotinylated ligand (EC₅₀: 0.3101–0.5710 ng/mL) .

  2. Receptor Interaction Studies:

    • Investigates binding to TNF-R1/R2 and downstream signaling pathways (e.g., NF-κB, JNK) .

  3. Cytotoxicity Assays:

    • Models TNF-α-mediated apoptosis in cancer cell lines (e.g., Du145 prostate cancer) .

Production and Quality Control

ParameterValue/DescriptionSource
EndotoxinNot tested (focus on bioactivity)
StorageLyophilized powder (reconstitution in sterile buffer recommended)
StabilityStable at 4°C for short-term storage; -20°C for long-term

The protein is validated for purity (>90%) via SDS-PAGE and bioactivity via cytotoxicity assays .

Comparative Analysis with Full-Length TNF-α

FeatureFull-Length TNF-αPartial TNF (77–233aa)
Molecular Weight26 kDa (monomer), 55 kDa (trimer) ~17 kDa (estimated) + His-tag
Receptor BindingHigh-affinity trimer binding (KD ~90 pM) Monomeric binding (affinity not quantified)
BioactivityED₅₀: 25–100 pg/mL (L-929 cells) ED₅₀: 33–47 pg/mL
Clinical RelevanceUsed in hyperthermic limb perfusion for sarcomas Research-focused (e.g., ELISA standards)

Case Study: Functional ELISA Application

A user demonstrated the partial TNF’s effectiveness as an ELISA standard when coated on plates and tested with TNFSF13B-Biotinylated ligand. The EC₅₀ of 0.3101–0.5710 ng/mL confirmed its utility in high-throughput screening for TNF-related ligands .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered phosphate-buffered saline (PBS), pH 7.0.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Avoid repeated freeze-thaw cycles. Store reconstituted aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to pellet the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting to -20°C/-80°C. Our standard protocol utilizes 50% glycerol. This concentration should serve as a guideline for customers.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is crucial for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
N-terminal 6xHis-tagged
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
18.3 kDa
Protein Length
Partial
Purity
>97% 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. Primarily secreted by macrophages, it can induce apoptosis in certain tumor cell lines. A potent pyrogen, TNF induces fever directly or by stimulating interleukin-1 secretion. It is also implicated in cachexia. However, under specific conditions, it can stimulate cell proliferation and differentiation. In individuals with rheumatoid arthritis, TNF impairs regulatory T-cell (Treg) function by dephosphorylating FOXP3. This upregulates protein phosphatase 1 (PP1), which dephosphorylates the crucial Ser-418 residue of FOXP3, thus inactivating it and rendering Tregs functionally defective. TNF is a key mediator of cell death in the anti-cancer effects of BCG-stimulated neutrophils in combination with DIABLO/SMAC mimetics in the RT4v6 bladder cancer cell line. It induces insulin resistance in adipocytes by inhibiting insulin-induced IRS1 tyrosine phosphorylation and glucose uptake, partly through inducing GKAP42 protein degradation. TNF also plays a role in angiogenesis by synergistically inducing VEGF production with IL1B and IL6. The intracellular domain (ICD) form of TNF induces IL12 production in dendritic cells.
Gene References Into Functions
  • Genetic predisposition to rheumatoid arthritis in the Russian population of the Republic of Karelia is associated with the GG TNF-alpha genotype. PMID: 30225702
  • Treatment with 30 µg/ml curcumin significantly reduces TNFalpha protein production in Behcet's disease patients (p < 0.01) and healthy controls (p < 0.05) M1 macrophages. PMID: 29806793
  • The A allele of TNF-alpha 238A/G, but not TNF-alpha 308A/G, shows a protective effect against juvenile idiopathic arthritis in the Caucasian population (Meta-Analysis). PMID: 30412082
  • Regionally expressed TNF-alpha in epicardial adipose tissue may strongly influence the progression of coronary atherosclerosis. PMID: 28931782
  • The effect of a pro-inflammatory diet on TNF-alpha concentrations is more pronounced in pregnant women reporting higher stress levels. PMID: 30200631
  • The TNF-alpha -308G/A polymorphism is not associated with the risk of bullous pemphigoid and alopecia areata in an Iranian cohort. PMID: 29843231
  • Only the promoter single nucleotide polymorphism (SNP) rs1800629 within the TNF-alpha gene is associated with an increased risk of Graves' disease (GD), particularly in European populations. Larger studies are needed to confirm these associations. PMID: 29440561
  • 173 polymorphisms were studied to determine their association with the response to anti-TNF drugs in patients with moderate-to-severe plaque psoriasis (N=144). PMID: 27670765
  • The SNP rs4819554 in the IL17RA promoter region significantly influences the response to anti-TNF drugs at week 12. PMID: 27670766
  • TNF-alpha promoter gene polymorphisms and/or haplotypes are risk factors for nephrotic syndrome and steroid resistance among Egyptian children. PMID: 28803697
  • The TNF-alpha gene polymorphism might be a risk factor for nasal polyposis in northern Iran; the minor frequency of the G308A allele is slightly higher than in other major populations. PMID: 30003390
  • The interplay of IFN-gamma (from CD4+ T lymphocytes) and TNF-alpha (from CD14+ cells) does not directly affect parasite replication but induces IL-4 production. PMID: 29953494
  • The single nucleotide polymorphisms rs361525, rs1800629, and rs17999645 of tumor necrosis factor-alpha were significantly correlated with the diagnosis of cervical cancer. PMID: 29940817
  • Rs11541076 in IRAK3, a negative regulator of TLR signaling, is a predictor of anti-TNF treatment response. PMID: 27698401
  • In a Brazilian population, TNF and IL17 gene polymorphisms associated with the expression of important inflammatory cytokines were associated with overall spondyloarthritis and specifically with ankylosing spondylitis and psoriatic arthritis, regardless of gender and HLA-B27. PMID: 29849482
  • By restraining TNFR1 at the cell surface via sialylation, ST6Gal-I acts as a functional switch, diverting signaling toward survival. This reveals a novel glycosylation-dependent mechanism regulating cellular response to TNF and potentially promoting cancer cell survival in TNF-rich tumor microenvironments. PMID: 29233887
  • The TNF*A allele confers susceptibility to autoimmune hepatitis (AIH) in Tunisian patients and is associated with increased TNF-alpha production. Anti-TNF antibodies could be an alternative to corticotherapy, avoiding exacerbated immune responses in AIH. PMID: 29845365
  • Impaired TNF, IL-1beta, and IL-17 production upon stimulation with mycobacterial antigens may contribute to increased susceptibility to M. tuberculosis infection in HTLV-1-infected individuals. PMID: 29523325
  • 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
  • The TNF-alpha -308G>A polymorphism affects the overall survival of cancer patients and is a potential therapeutic target. PMID: 30407345
  • Many inflammatory pathologies are driven by aberrant TNF-induced cell death, which mostly depends on the kinase RIPK1 (Receptor-interacting serine/threonine-protein kinase 1). [review] PMID: 29217118
  • High TNF-alpha expression is associated with retinopathy of prematurity. PMID: 29274846
  • TNF-alpha selectively reduces BMPR-II transcription and mediates post-translational BMPR-II cleavage via ADAM10 and ADAM17 in pulmonary artery smooth muscle cells. PMID: 28084316
  • Polymorphisms of IL-1betab and TNF-a are not a risk factor for intrahepatic cholangiocarcinoma (ICC), but O. viverrini infection affects all TNF-alpha genotypes, potentially promoting ICC. Primary ICC prevention in high-risk areas focuses on reducing O. viverrini infection. PMID: 30139338
  • In the placenta, controlling for gestational age, TNF-alpha and MMP-9 showed a significant positive correlation only in the second trimester. PMID: 28820024
  • In human endometrial stromal cells, high TNF levels negatively affect insulin action through decreased adiponectin signaling and GLUT4 protein. This may explain impaired endometrial function in obese women with polycystic ovary syndrome. PMID: 28946816
  • Three SNPs in P2X4R and two in CAMKK2 influenced TNFalpha concentrations in peripheral blood mononuclear cells but did not associate with HIV-associated sensory neuropathy risk in South Africans. PMID: 29428485
  • TNF-alpha impairs endothelial tight junctions and promotes monocyte-endothelial cell adhesion by upregulating BACE1 expression, activating PKC signaling, and subsequently cleaving ST6Gal1. PMID: 28091531
  • PrP enhances responses to TNF-alpha, promoting proinflammatory cytokine production, which may contribute to inflammation and tumorigenesis. PMID: 28900035
  • The protein and mRNA expression levels of the cytokine TNF-alpha are significantly increased. PMID: 28476335
  • Wnt/beta-catenin signal pathway activation-dependent upregulation of syncytin-1 contributes to TNF-alpha-enhanced fusion between oral squamous cell carcinoma cells and endothelial cells. PMID: 28112190
  • In children with pediatric obesity, lifestyle weight-loss intervention downregulates serum cardiotrophin-1 (CTF1), interleukin-6 (IL6), and tumor necrosis factor-alpha (TNFA); expression is also downregulated in peripheral blood mononuclear cells after improvement in adiposity, BMI, and waist-hip ratio. PMID: 28749076
  • TNFalpha differently regulated the levels of PPARalpha, LXRalpha, and LXRbeta binding to the apoA-I gene promoter in THP-1 cells. This suggests a novel tissue-specific mechanism of TNFalpha-mediated regulation of apoA-I in monocytes and macrophages, showing that endogenous ApoA-I might be positively regulated in macrophages during inflammation. PMID: 29442267
  • TET2 mutations promote clonal dominance with aging by conferring TNFalpha resistance to sensitive bone marrow progenitors and propagating an inflammatory environment. PMID: 29195897
  • The anti-rotavirus effect of TNF-alpha is achieved by NFkappaB-regulated genes via activation of classical NF-kappaB signaling. PMID: 29859235
  • The heterozygous genotype GA of TNF-alpha-238 (rs 361525) SNP significantly increased the risk of adverse outcomes (mortality rate), regardless of organ dysfunction or sepsis severity. PMID: 29978383
  • In patients with primary depression, depressive symptoms were associated with TNF-alpha. PMID: 30148175
  • TNFalpha causes CD80 upregulation, actin reorganization, and podocyte injury in podocytes. PMID: 29022109
  • C-reactive protein and TNF-alpha levels are important diagnostic markers of inflammation in patients with chronic pancreatitis and type 2 diabetes mellitus. PMID: 30280549
  • TNF-alpha and IL-10 treatment affects ICAM-1 and CD31 expression in human coronary artery endothelial cells. PMID: 29949812
  • KLF15 overexpression in Eahy926 cells protects against TNFalpha-induced dysfunction by activating Nrf2 signaling and inhibiting NF-kappaB signaling. PMID: 29956764
  • 30 and 100 ng/ml TIMP3 attenuate migration and proliferation, and inhibit MMP2, MMP9, and TNFalpha secretion of NA SMCs. TIMP3 may be a potential therapeutic drug in novel drug-eluting stents to attenuate progressive aortic NA dilation. PMID: 29956789
  • Elevated A20 promotes TNF-induced and RIPK1-dependent intestinal epithelial cell death. PMID: 30209212
  • TNF-alpha GG genotype at -238 and GG haplotype at -308 and -238 were associated with Kawasaki disease in an Iranian population. PMID: 27455075
  • A TNFalpha gene polymorphism, rs1800629, is highly significantly associated with postmenopausal osteoporosis and BMD in female Han Chinese populations. PMID: 29481288
  • The -308A allele of TNF-alpha may play a role in rheumatoid arthritis progression in a North Indian population. PMID: 28748515
  • ATF3 mediates the inhibitory action of TNF-alpha on osteoblast differentiation; the TNF-alpha-activated JNK pathway induces Atf3 expression. PMID: 29605296
  • TNF-alpha, DKK1, and OPG are involved in knee osteoarthritis pathogenesis. PMID: 28676900
  • 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
  • Prolonged TNFalpha exposure is detrimental to endothelial cells, causing senescence; chronically increased TNFalpha levels may contribute to chronic inflammatory disease pathology by driving premature endothelial senescence. PMID: 28045034
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.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Flow Cytometry

Review: A proliferation analysis was carried out using fluorescence microscopy and flow cytometry. Cell proliferation was analyzed at 12, 24 and 36 hours with no stimulation (control), stimulate with TNF or stimulated with TNF + CS.

Q&A

What is the active structural form of recombinant human TNF?

Recombinant human TNF functions as a trimer with a molecular weight of approximately 55,000 Daltons. This trimeric structure is essential for optimal biological activity, as monomeric TNF (approximately 17,000 Daltons) exhibits significantly reduced binding affinity and cytotoxicity. Studies have demonstrated that the trimeric form shows at least 8-fold higher activity than monomeric TNF, with high binding affinity (KD = 90 pM) compared to monomers (KD = 70 nM). The trimeric conformation creates three receptor binding sites, formed as grooves between adjacent subunits, enabling efficient receptor clustering and subsequent signal transduction .

How stable is the trimeric form of recombinant TNF in experimental conditions?

The stability of TNF trimers depends significantly on experimental conditions. The presence of detergents like Triton X-100 can partially dissociate the human TNF trimer into monomers with consequent reduction in activity. For maintaining trimeric stability, researchers should avoid detergents when possible and use appropriate buffer systems. Cross-linking agents such as bis[2-(succinimidooxycarbonyloxy)ethyl]sulfone have been employed to stabilize trimers for certain experimental applications. Temperature fluctuations and freeze-thaw cycles should be minimized, as these can compromise the quaternary structure of the protein and reduce biological activity .

What methodologies can confirm the trimeric state of recombinant TNF preparations?

Verification of the trimeric state of recombinant TNF can be accomplished through multiple complementary techniques:

  • Gel filtration chromatography on columns such as Sephadex G-75, which can separate the ~55,000 Da trimers from ~17,000 Da monomers

  • Native PAGE electrophoresis under non-denaturing conditions

  • Dynamic light scattering (DLS) to assess size distribution

  • Analytical ultracentrifugation for sedimentation analysis

  • Functional assays comparing binding affinity to TNF receptors, as trimers demonstrate significantly higher binding affinity (KD = 90 pM) than monomers (KD = 70 nM)

These analyses should be conducted before experimental use to ensure consistency in biological activity across preparations .

How do the binding characteristics differ between TNFR1 and TNFR2?

TNFR1 (p55) and TNFR2 (p75) exhibit notable differences in their TNF binding characteristics:

ParameterTNFR1TNFR2
Binding affinity for TNFHigher (KD = 1.9 × 10^-11 M)Lower (KD = 4.2 × 10^-10 M)
Complex stabilityStable, long-livedShort-lived, transient
Response to soluble TNFRobust activationWeak activation
Response to transmembrane TNFActivationRobust activation
Mechanical requirementsSimple binding sufficientRequires cluster formation

These differences significantly impact experimental design choices when studying receptor-specific functions. TNFR2 activation particularly requires either transmembrane TNF or artificially clustered TNF to create stable signaling complexes, whereas TNFR1 can be effectively activated by soluble TNF alone .

What experimental approaches can distinguish between TNFR1 and TNFR2-mediated effects?

To differentiate between TNFR1 and TNFR2-mediated effects in research settings, the following methodological approaches are recommended:

  • Receptor-selective TNF muteins: Use engineered TNF variants such as R1antTNF that selectively bind TNFR1 without activating TNFR2 signaling, or TNFR2-selective agonists

  • Receptor-specific antibodies: Employ antagonistic or agonistic antibodies targeting specific receptors

  • Receptor knockout models: Utilize cell lines or animal models with genetic deletion of either TNFR1 or TNFR2

  • Differential ligand presentation: Implement immobilized versus soluble TNF presentations, as immobilized TNF better activates TNFR2

  • Selective inhibition of downstream pathways: Target TNFR1-specific (primarily apoptotic) versus TNFR2-specific (primarily proliferative) signaling components

These approaches enable precise delineation of receptor-specific functions in complex biological systems and can reveal differential roles in disease pathogenesis .

How does transmembrane versus soluble TNF influence receptor activation patterns?

Transmembrane TNF (tmTNF) and soluble TNF (sTNF) exhibit distinct receptor activation profiles that significantly impact experimental outcomes. sTNF efficiently activates TNFR1 but provides only weak stimulation of TNFR2, despite binding to TNFR2 with subnanomolar affinity. This differential activation stems from the association/dissociation kinetics of the TNF-receptor complexes. While sTNF forms stable, signaling-competent complexes with TNFR1, it creates only transient, signaling-incompetent complexes with TNFR2. In contrast, tmTNF robustly activates both receptors.

Studies using nanoscale patterning systems demonstrated that mechanical fixation of TNF alone was sufficient for TNFR1 activation but inadequate for TNFR2 activation, which required additional stabilization through cluster formation. These findings highlight the importance of ligand presentation in experimental design, particularly when studying TNFR2-mediated processes .

What are optimal reconstitution protocols for lyophilized recombinant TNF?

Reconstitution of lyophilized recombinant human TNF requires careful attention to maintain structural integrity and biological activity. The recommended procedure involves:

  • Allow the vial to reach room temperature before opening

  • Reconstitute with sterile distilled water (typically 0.1 ml for standard preparations)

  • Gently mix rather than vortex, as the protein may appear as a film at the bottom of the vial

  • Allow complete dissolution before aliquoting to avoid heterogeneity

  • For long-term storage, addition of 0.09% sodium azide is recommended, but importantly, sodium azide should be omitted for functional studies as it may interfere with biological activity

The reconstituted protein should be stored at -20°C or -80°C in single-use aliquots to avoid repeated freeze-thaw cycles. Always verify protein concentration after reconstitution using standard protein assays .

What quality control measures ensure consistent activity of recombinant TNF preparations?

To ensure experimental reproducibility, recombinant TNF preparations should undergo rigorous quality control testing:

  • Purity assessment: SDS-PAGE analysis (>95% purity expected for research-grade material)

  • Endotoxin testing: Limulus Amebocyte Lysate (LAL) assay to confirm levels below 0.1 EU/μg protein

  • Structural verification: Size exclusion chromatography to confirm trimeric state

  • Biological activity assays:

    • Cytotoxicity assay using L929 fibroblasts in the presence of actinomycin D

    • Receptor binding assays with HeLa cells or receptor-expressing cell lines

    • NF-κB activation reporter assays

  • Batch-to-batch consistency verification through standardized reference preparations

Researchers should maintain detailed records of these quality control parameters for each preparation and include them in experimental documentation to ensure reproducibility across studies .

How should researchers determine optimal TNF concentrations for specific cell types?

Determination of optimal TNF concentrations requires systematic titration experiments tailored to the specific experimental system:

  • Conduct initial dose-response studies covering a broad concentration range (typically 0.1-100 ng/ml)

  • Evaluate multiple relevant readouts, including:

    • Cell viability and proliferation

    • Receptor signaling activation (phosphorylation of downstream mediators)

    • Target gene expression

    • Protein secretion profiles

    • Cell surface marker modulation

  • Establish time-course experiments to determine both acute and sustained responses

  • Consider cell type-specific sensitivity variations; immune cells often respond to lower concentrations than epithelial or stromal cells

  • For co-culture systems, determine concentrations that appropriately activate the primary cell type without excessive off-target effects on secondary cell populations

These systematic approaches prevent experimental artifacts from inappropriate dosing and ensure physiologically relevant responses. The biological potency should be reported in units of activity rather than simple concentration whenever possible .

What cell-based assays effectively measure TNF bioactivity?

Several established bioassays can quantitatively assess TNF activity in experimental preparations:

  • L929 mouse fibroblast cytotoxicity assay:

    • Cells are sensitized with actinomycin D (1 μg/ml)

    • Serial dilutions of TNF are applied for 18-24 hours

    • Cell viability is assessed via MTT or neutral red uptake

    • EC50 values are calculated to determine potency

  • NF-κB reporter assays:

    • Cells expressing luciferase or GFP under NF-κB responsive elements

    • Quantification of reporter activation following TNF treatment

    • Allows measurement of early signaling events

  • Receptor binding assays:

    • Using cells expressing TNFR1 or TNFR2 (HeLa cells express both naturally)

    • Competitive binding with labeled TNF preparations

    • Determination of binding constants and receptor occupancy

  • JNK/p38 MAPK phosphorylation:

    • Western blot or flow cytometry-based detection of phosphorylated signaling proteins

    • Provides quantitative assessment of early TNF-induced signaling

These assays should include appropriate positive controls and reference standards to enable comparisons across different experimental settings .

How can researchers distinguish between TNF-induced apoptosis and necroptosis experimentally?

Distinguishing between TNF-induced apoptosis and necroptosis requires specific methodological approaches:

  • Inhibitor studies:

    • Z-VAD-FMK (pan-caspase inhibitor): Blocks apoptosis but can enhance necroptosis

    • Necrostatin-1 (RIPK1 inhibitor): Specifically blocks necroptosis

    • Combined treatment can differentiate between cell death modalities

  • Molecular markers:

    • Apoptosis: Caspase-3/7 activation, PARP cleavage, phosphatidylserine externalization (Annexin V+/PI-)

    • Necroptosis: MLKL phosphorylation, membrane permeabilization without prior caspase activation (PI+ without Annexin V single-positive stage)

  • Morphological assessment:

    • Apoptosis: Nuclear fragmentation, membrane blebbing, cellular shrinkage

    • Necroptosis: Cellular swelling, rapid membrane permeabilization, organelle swelling

  • Genetic approaches:

    • siRNA knockdown or CRISPR/Cas9 deletion of key pathway components (caspases for apoptosis; RIPK1, RIPK3, or MLKL for necroptosis)

    • Reconstitution experiments to confirm specificity

Combining multiple approaches provides the most reliable differentiation between these cell death pathways in TNF-stimulated experimental systems .

How can recombinant TNF variants be designed for receptor-selective signaling studies?

Engineering receptor-selective TNF variants involves strategic molecular modifications based on structural understanding:

  • TNFR1-selective antagonists:

    • The R1antTNF variant (mutations: A84S, V85T, S86T, Y87H, Q88N, T89Q) selectively binds TNFR1 without activating signaling

    • The critical Y87H mutation changes binding from hydrophobic to electrostatic interaction, creating unstable TNFR1 binding that prevents signal transduction

    • Further stability improvements can be achieved through:

      • PEGylation to extend half-life (PEG-R1antTNF)

      • Single-chain derivatives with peptide linkers between protomers (scR1antTNF)

  • TNFR2-selective agonists:

    • Mutations enhancing TNFR2 binding while reducing TNFR1 affinity

    • Oligomerization strategies to promote TNFR2 clustering required for signaling

    • Domain fusion approaches combining TNF with multimerization domains

  • Functional validation approaches:

    • Receptor binding assays comparing wild-type and mutant TNF

    • Pathway-specific readouts (NF-κB for inflammatory signaling; caspase activation for apoptosis)

    • Animal models examining efficacy and specificity in vivo

These engineered variants enable precise dissection of receptor-specific functions in complex disease models and may represent promising therapeutic approaches with reduced side effects compared to global TNF inhibition .

What are methodological considerations for using TNF in disease model systems?

Implementing TNF in disease model systems requires careful methodological planning:

  • Dose determination:

    • Use physiologically relevant concentrations based on measurements from patient samples (typically 1-100 pg/ml in serum during inflammatory conditions)

    • Establish dose-response relationships specific to the model system

    • Consider local versus systemic concentrations (local tissue concentrations may be 10-100× higher than serum)

  • Timing considerations:

    • Acute versus chronic administration protocols

    • Pulsatile versus continuous exposure

    • Pre-conditioning versus therapeutic intervention timing

  • Delivery methods:

    • Local administration for tissue-specific effects

    • Osmotic pumps for continuous delivery

    • Inducible expression systems for regulated production

    • Cell-specific targeting approaches

  • Complementary approaches:

    • Combine with TNF receptor knockout models to determine receptor specificity

    • Use TNF-deficient backgrounds for add-back experiments

    • Compare transmembrane-restricted TNF (non-cleavable mutants) versus wild-type TNF

    • Consider combination with other cytokines relevant to the disease context

  • Translational considerations:

    • Species-specific differences in TNF biology and receptor distribution

    • Human versus mouse TNF specificity (human TNF poorly activates mouse TNFR1)

    • Need for humanized models in certain therapeutic applications

These methodological considerations ensure that TNF-based disease models accurately recapitulate human pathophysiology and provide reliable preclinical data for therapeutic development .

How do current approaches for selective TNF receptor targeting differ from global TNF inhibition?

Selective TNF receptor targeting represents an advanced approach compared to global TNF inhibition, with distinct methodological implementations:

ApproachMechanismExamplesMethodological Considerations
Global TNF inhibitionNeutralizes all TNF activity regardless of receptorAnti-TNF antibodies (infliximab), soluble TNF receptors (etanercept)Complete blockade of both beneficial and harmful TNF effects; associated with infection risk and paradoxical inflammation
TNFR1-selective inhibitionBlocks pro-inflammatory and apoptotic signaling while preserving TNFR2 immunoregulatory functionsR1antTNF muteins, TNFR1-selective antibodiesRequires high specificity validation; potential to reduce side effects while maintaining efficacy; preservation of host defense mechanisms
TNFR2-selective stimulationEnhances immunoregulatory and tissue-regenerative functions without activating TNFR1 inflammatory pathwaysTNFR2-selective agonistic antibodies, engineered TNFR2-specific ligandsRequires careful dosing to avoid off-target effects; potential for both immunosuppressive and regenerative applications

Experimental approaches studying selective receptor targeting require:

  • Rigorous validation of receptor selectivity through binding and functional assays

  • Comprehensive comparison with global TNF inhibition in parallel experiments

  • Monitoring of both intended therapeutic effects and potential adverse outcomes

  • Assessment of immunological competence during treatment, particularly for antimicrobial responses

Current research indicates selective targeting approaches may offer superior therapeutic indices for certain inflammatory and autoimmune conditions while mitigating side effects associated with global TNF blockade .

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.