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.
Parameter | Value/Description | Source |
---|---|---|
Cytotoxicity (ED₅₀) | 33.32–47.38 pg/mL (L-929 mouse fibroblasts + actinomycin D) | |
Receptor Binding | Binds 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 .
ELISA Standard:
Receptor Interaction Studies:
Cytotoxicity Assays:
The protein is validated for purity (>90%) via SDS-PAGE and bioactivity via cytotoxicity assays .
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 .
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.
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 .
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 .
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 .
TNFR1 (p55) and TNFR2 (p75) exhibit notable differences in their TNF binding characteristics:
Parameter | TNFR1 | TNFR2 |
---|---|---|
Binding affinity for TNF | Higher (KD = 1.9 × 10^-11 M) | Lower (KD = 4.2 × 10^-10 M) |
Complex stability | Stable, long-lived | Short-lived, transient |
Response to soluble TNF | Robust activation | Weak activation |
Response to transmembrane TNF | Activation | Robust activation |
Mechanical requirements | Simple binding sufficient | Requires 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 .
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 .
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 .
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 .
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 .
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 .
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 .
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 .
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 .
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 .
Selective TNF receptor targeting represents an advanced approach compared to global TNF inhibition, with distinct methodological implementations:
Approach | Mechanism | Examples | Methodological Considerations |
---|---|---|---|
Global TNF inhibition | Neutralizes all TNF activity regardless of receptor | Anti-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 inhibition | Blocks pro-inflammatory and apoptotic signaling while preserving TNFR2 immunoregulatory functions | R1antTNF muteins, TNFR1-selective antibodies | Requires high specificity validation; potential to reduce side effects while maintaining efficacy; preservation of host defense mechanisms |
TNFR2-selective stimulation | Enhances immunoregulatory and tissue-regenerative functions without activating TNFR1 inflammatory pathways | TNFR2-selective agonistic antibodies, engineered TNFR2-specific ligands | Requires 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 .