Tumor Necrosis Factor-α (TNF-α): A Central Mediator of Inflammation and Immune Regulation
Molecular Structure and Biosynthetic Characteristics
Tumor Necrosis Factor-α (TNF-α) is a 233-amino acid type II transmembrane protein that, upon cleavage by TNF-α-converting enzyme (TACE/ADAM17), releases a soluble, biologically active 157-amino acid form (17 kDa). Structurally, TNF-α exists as a stable homotrimer, with each monomer featuring a characteristic "TNF fold" composed of two antiparallel β-sheets. X-ray crystallography reveals that this trimeric conformation induces specific activating conformations in TNFR1 or TNFR2 upon binding. The TNF-α gene (TNFSF2), located on the short arm of human chromosome 6 (6p21.3), is tightly regulated: its promoter region contains binding sites for transcription factors such as NF-κB, AP-1, and CREB, and in macrophages, TNF-α mRNA levels can surge 50-100-fold within 30 minutes of LPS stimulation. Notably, TNF-α biosynthesis is controlled at multiple levels: mRNA stability is regulated by AU-rich elements (AREs) in the 3' untranslated region (3'UTR) interacting with RNA-binding proteins like tristetraprolin (TTP); translation is controlled via an internal ribosome entry site (IRES); and secretion depends on the proteolytic activity of ADAM17. Under normal physiological conditions, serum TNF-α levels remain very low (<10 pg/mL), but during systemic inflammatory responses like sepsis, they can skyrocket to over 1000 pg/mL.

Receptor System and Signal Transduction Mechanisms
TNF-α signals through two transmembrane receptors, TNFR1 (p55/CD120a) and TNFR2 (p75/CD120b), forming a complex cellular response network. TNFR1 is ubiquitously expressed in nearly all nucleated cells and exhibits biphasic signaling: initially, it activates NF-κB and MAPK pathways via the TRADD/RIP1/TRAF2 complex to promote inflammatory cytokine expression and cell survival; later, it induces apoptosis through FADD/caspase-8. This time-dependent functional switch is finely regulated by the subcellular localization and post-translational modifications of receptor complexes, particularly the ubiquitination status of RIP1, which determines signaling outcomes. In contrast, TNFR2 expression is more tissue-specific (primarily in immune cells, endothelial cells, and neurons) and directly activates PI3K/Akt and NF-κB pathways via TRAF2, playing a key role in tissue repair and immune regulation. Recent studies show that soluble TNF-α (sTNF-α) primarily activates TNFR1-mediated pro-inflammatory responses, while membrane-bound TNF-α (tmTNF-α) more effectively triggers TNFR2-mediated pro-survival signals. Regulatory molecules like the deubiquitinase A20 and the linear ubiquitin chain assembly complex (LUBAC) act as "molecular brakes" to prevent excessive signaling. Notably, TNFR1 and TNFR2 cross-regulate each other, with TNFR2 inhibiting TNFR1 signaling by sequestering TRAF2, adding complexity to the signaling network.
Central Role in Inflammatory Responses
As a key initiator of inflammatory cascades, TNF-α plays a pivotal role in acute and chronic inflammation. Upon recognition of pathogen-associated molecular patterns (PAMPs) or tissue damage signals, TNF-α triggers inflammation through three mechanisms: upregulating endothelial adhesion molecules (ICAM-1, VCAM-1, and E-selectin) to promote leukocyte rolling, adhesion, and transendothelial migration; stimulating hepatocytes to produce acute-phase proteins (e.g., C-reactive protein and serum amyloid A); and activating fibroblasts and epithelial cells to release chemokines (IL-8, MCP-1) and other pro-inflammatory cytokines (IL-1β, IL-6). At the molecular level, TNF-α induces the expression of inflammatory mediators via NF-κB, including cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and matrix metalloproteinases (MMP-1/3/9). Animal studies demonstrate that TNF-α knockout mice are completely resistant to LPS-induced endotoxic shock but exhibit impaired clearance of intracellular pathogens like Listeria. In chronic inflammation, persistent TNF-α drives inflammatory cell infiltration and tissue remodeling, such as synovial hyperplasia and bone erosion in rheumatoid arthritis (RA). Clinically, serum TNF-α levels correlate with disease activity in various inflammatory conditions, reaching over 100-fold higher in sepsis patients than in healthy individuals. Interestingly, TNF-α also participates in inflammation resolution by inducing anti-inflammatory factors (e.g., IL-10) and promoting macrophage phenotype switching, a bidirectional regulatory function critical for immune homeostasis.
Pathological Mechanisms in Autoimmune Diseases
Dysregulated TNF-α expression is closely linked to the pathogenesis and progression of autoimmune diseases, making it a key therapeutic target. In RA, synovial fluid TNF-α concentrations can be 100-fold higher than in healthy individuals, driving joint destruction through multiple mechanisms: enhancing osteoclast differentiation and bone resorption (5-10-fold increase in RANKL expression); inhibiting chondrocyte synthesis of proteoglycans; and inducing synovial fibroblasts to produce MMP-3 and MMP-13, which degrade cartilage matrix. Genome-wide association studies (GWAS) identify the -308G>A (rs1800629) polymorphism in the TNF promoter as a risk factor for RA, with the A allele increasing TNF-α transcriptional activity 2-3-fold. In ankylosing spondylitis (AS), tmTNF-α activates osteogenic differentiation of mesenchymal stem cells via TNFR2, leading to ligament ossification and spinal fusion. In inflammatory bowel disease (IBD), TNF-α increases intestinal epithelial permeability (60-80% reduction in transepithelial resistance), disrupts tight junction proteins (e.g., occludin and claudins), and promotes Th1/Th17 responses. Systemic lupus erythematosus (SLE) patients show elevated serum TNF-α levels correlating with disease activity (SLEDAI), though paradoxically, TNF-α may have protective effects in some SLE models. In multiple sclerosis (MS), TNF-α contributes to central nervous system inflammation by disrupting the blood-brain barrier, yet anti-TNF-α therapy unexpectedly worsens symptoms, highlighting its spatiotemporal complexity.
Clinical Applications of Anti-TNF-α Therapy
Anti-TNF-α therapy has revolutionized autoimmune disease treatment, with over 7 million patients worldwide receiving these biologics. Clinically used anti-TNF-α agents fall into three categories: monoclonal antibodies (e.g., adalimumab, infliximab), receptor fusion proteins (etanercept), and PEGylated Fab fragments (certolizumab pegol). These drugs differ pharmacokinetically and pharmacodynamically: IgG1 monoclonal antibodies (e.g., adalimumab) neutralize both soluble and membrane-bound TNF-α, have a ~2-week half-life, and mediate antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) via Fc regions; etanercept (a TNFR2-Fc fusion) primarily neutralizes soluble TNF-α with a shorter half-life (3-5 days). Clinical trials show anti-TNF-α therapy improves ACR50 response rates in RA from ~10% (placebo) to 50-60%, with >70% reduction in radiographic progression. In Crohn’s disease, 60-70% of patients achieve clinical remission. However, 30-40% experience treatment failure due to anti-drug antibodies (5-15% incidence), alternative inflammatory pathway activation, or epigenetic remodeling. Next-generation strategies include nanobodies for better tissue penetration, bispecific antibodies targeting TNF-α and IL-17/IL-23, and prodrug antibodies activated selectively in inflammatory microenvironments. Therapeutic drug monitoring (serum drug levels and anti-drug antibodies) guided dose optimization improves response rates by 15-20%.
Dual Roles in Infection and Tumor Immunity
TNF-α exhibits context-dependent roles in infection defense and tumor immunity. In bacterial infections, it enhances macrophage and neutrophil bactericidal activity and promotes granuloma formation to contain pathogens. Anti-TNF-α therapy increases latent tuberculosis reactivation risk 4-10-fold. In viral infections, TNF-α limits replication by inducing apoptosis, though viruses like EBV and HCV evade its signaling. In cancer, TNF-α has dose-dependent biphasic effects: physiological levels activate endothelial cells and cytotoxic T cells for immune surveillance, while chronic overproduction creates a pro-tumor microenvironment. Anti-TNF-α therapy slightly increases lymphoma risk (standardized incidence ratio [SIR]=1.5-3.0). Notably, local high-dose TNF-α selectively disrupts tumor vasculature, achieving 60-80% response rates in melanoma and sarcoma. Combining TNF-α blockade with PD-1 inhibitors may enhance efficacy, a hypothesis under clinical investigation.
Future Directions and Therapeutic Prospects
TNF-α research is advancing toward precision and medicine. Single-cell multi-omics reveal cell-specific TNF-α functions, informing targeted strategies. Epigenetic studies link TNF locus DNA methylation to treatment response, enabling personalized therapy. Nanomedicine innovations include pH-responsive TNF-α inhibitors for site-specific intervention. Bispecific molecules like TNF-α/IL-23 nanobodies show superior efficacy in psoriasis models. CRISPR-Cas9-engineered TNF-α reporter models facilitate high-throughput drug screening. Clinical applications are expanding to Alzheimer’s disease (neuroinflammation) and metabolic syndrome (chronic low-grade inflammation). In the next 5 years, 10-15 novel TNF-α modulators are expected to enter clinical trials, with AI-guided personalized regimens ushering in a new era of precision medicine. As understanding of TNF-α signaling deepens, selective targeting of specific receptors or downstream pathways may improve efficacy while minimizing adverse effects, offering breakthroughs for treating major diseases.
Click on the product catalog numbers below to access detailed information on our official website.
Product Information
|
Human TNF-α Surpass ELISA PairSet Kit |
||
|
TNF-α Protein, Mouse |
Host : Mouse Expression System : E.coli Conjugation : Unconjugated |
|
|
TNF-α Protein, Human |
Host : Human Expression System : CHO Conjugation : Unconjugated |
|
|
TNF-α Protein, Human |
Host : Human Expression System : E.coli Conjugation : Unconjugated |
|
|
TNF-α Protein, Human |
Host : Human Expression System : E.coli Conjugation : Unconjugated |
|
|
TNF-α Protein, Rat |
Host : Rat Expression System : E.coli Conjugation : Unconjugated |
|
|
Human TNF-α OneStep ELISA Kit |
||
|
TNF-α Protein, Mouse |
Host : Mouse Expression System : E.coli Conjugation : Unconjugated |
|
|
Mouse TNF-α OneStep ELISA Kit |