Although activity that induced tumor regression was observed and termed tumor necrosis factor (TNF) as early as the 1960s, the true identity of TNF was not clear until 1984, when Aggarwal and coworkers reported, for the first time, the isolation of 2 cytotoxic factors: one, derived from macrophages (molecular mass 17 kDa), was named TNF, and the second, derived from lymphocytes (20 kDa), was named lymphotoxin. Because the 2 cytotoxic factors exhibited 50% amino acid sequence homology and bound to the same receptor, they came to be called TNF-α and TNF-β. Identification of the protein sequences led to cloning of their cDNA. Based on sequence homology to TNF-α, now a total of 19 members of the TNF superfamily have been identified, along with 29 interacting receptors, and several molecules that interact with the cytoplasmic domain of these receptors. The roles of the TNF superfamily in inflammation, apoptosis, proliferation, invasion, angiogenesis, metastasis, and morphogenesis have been documented. Their roles in immunologic, cardiovascular, neurologic, pulmonary, and metabolic diseases are becoming apparent. TNF superfamily members are active targets for drug development, as indicated by the recent approval and expanding market of TNF blockers used to treat rheumatoid arthritis, psoriasis, Crohns disease, and osteoporosis, with a total market of more than US $20 billion. As we learn more about this family, more therapeutics will probably emerge. In this review, we summarize the initial discovery of TNF-α, and the insights gained regarding the roles of this molecule and its related family members in normal physiology and disease.

The tumor necrosis factor (TNF) superfamily, composed of 19 ligands and 29 receptors, plays highly diversified roles in the body. The interest in TNF research has increased dramatically over the years as indicated by more than 113 000 citations on TNF-α alone, 27 000 on anti–TNF-α, 25 000 on TNF-α inhibitors, 55 000 on TNF receptors, 12 000 on TNF-mediated apoptosis, 40 000 on TNF-α-induced signals, and 9610 reviews. All members of the TNF superfamily, without exception, exhibit pro-inflammatory activity, in part through activation of the transcription factor NF-κB. Several members of the TNF superfamily exhibit proliferative activity on hematopoietic cells, in part through activation of various mitogen-activated kinases, and some members of this family play a role in apoptosis (Figure 1).1,2  Some members of the TNF superfamily have also been reported to play a role in morphogenetic changes and differentiation. Most members of the TNF superfamily have both beneficial and potentially harmful effects.3  Although TNF-α, for example, has been linked with physiologic proliferation and differentiation of B cells under steady-state conditions, it also has been linked with a wide variety of diseases, including cancer, cardiovascular, neurologic, pulmonary, autoimmune, and metabolic disorders.

Figure 1

Roles of various members of the TNF superfamily in inflammation, cellular proliferation, apoptosis, and morphogenesis. All members of the TNF superfamily exhibit pro-inflammatory activity, in part through activation of the transcription factor NF-κB (full red circle); OX40L, CD40L, CD27L, APRIL, and BAFF exhibit proliferative activity in part through activation of various mitogen-activated kinases (sky blue); TNF-α, TNF-β, FasL, and TRAIL control apoptosis (bluish-green); and EDA-A1, EDA-A2, TNF-α, FasL, and TRAIL regulate morphogenesis (green).

Figure 1

Roles of various members of the TNF superfamily in inflammation, cellular proliferation, apoptosis, and morphogenesis. All members of the TNF superfamily exhibit pro-inflammatory activity, in part through activation of the transcription factor NF-κB (full red circle); OX40L, CD40L, CD27L, APRIL, and BAFF exhibit proliferative activity in part through activation of various mitogen-activated kinases (sky blue); TNF-α, TNF-β, FasL, and TRAIL control apoptosis (bluish-green); and EDA-A1, EDA-A2, TNF-α, FasL, and TRAIL regulate morphogenesis (green).

Close modal

Although the fascinating history of TNF could be traced back more than one century, the name “tumor necrotizing factor” was first used in 1962 for tumor (sarcoma 37) regression activity induced in the serum of mice treated with Serratia marcescens polysaccharide; this activity was shown by Carswell et al in 1975 to be the result of TNF.4,5  Whether this TNF is the same as what our group later renamed TNF-α is unclear. In 1984 to 1985, our group structurally identified 2 different TNFs and cloned their genes. From hundreds of liters of conditioned medium collected from the human lymphoblastoid cell line RPMI 1788, we purified a protein 25 kDa in size, which initially was named lymphotoxin-α (LT-α) and later changed to TNF-β with the later discovery of sequence homology with TNF-α.6,7  Using the same cell lysis assays and antibodies against lymphotoxin, we then reported isolation of a second cytotoxic factor from hundreds of liters of conditioned supernatants of human promyelomonocytic cell line HL-60; this factor had a molecular mass approximately 17 kDa and was named human TNF-α.8  When we first performed the amino acid sequencing of the 2 proteins, it became clear that they exhibited as much as 50% sequence homology.6-8  Generation of antibodies against each molecule proved that these 2 proteins were immunologically distinct9 ; whereas TNF-α was produced by macrophages, TNF-β was produced by lymphocytes.10 

The amino acid sequence was used to prepare and isolate the full-length cDNAs for TNF-α and TNF-β.11,12  The cloning of the cDNA for TNF-β turned out to be much more difficult than that of TNF-α, for unclear reasons; thus, for TNF-β, most of the gene actually had to be chemically synthesized on the basis of the protein sequence.12  Soon our group discovered that both TNF-α and TNF-β bind to a common high-affinity cell surface receptor present on most cell types.13  While we were finishing the isolation of the TNF-α and TNF-β from monocytes and lymphocytes, respectively, Beutler et al from Rockefeller University reported the identity of TNF with the macrophage-secreted factor cachectin,14  which was responsible for cachexia. This protein was found to be a murine counterpart of human TNF-α, based on the amino acid sequence homology to human TNF-α. Cerami gave his account of this event in a recent article.15  Although passive immunization against cachectin/TNF has been shown to protect mice from the lethal effect of endotoxin, there are reports suggesting that endotoxin-induced factor is distinct from TNF-α.14 

More than 25 years later, 19 different members of the TNF superfamily have been identified based on their gene sequences (Figure 2; Table 1). TNF-α and TNF-β are the only members of this family that were first identified at the protein level, before isolation of their cDNA; all other members of this family were identified via human cDNA sequence homology. The superfamily members are TNF-α, TNF-β, lymphotoxin-β, CD40L, FasL, CD30L, 4-1BBL, CD27L, OX40L, TNF-related apoptosis-inducing ligand (TRAIL), LIGHT, receptor activator of NF-κB ligand (RANKL), TNF-related weak inducer of apoptosis (TWEAK), a proliferation-inducing ligand (APRIL), B-cell activating factor (BAFF), vascular endothelial cell-growth inhibitor (VEGI), ectodysplasin A (EDA)–A1, EDA-A2, and GITRL. These 19 members bind to 29 different receptors (Figure 2; Table 1). In addition, 3 TNF receptors (ie, TNFRSF22-mDcTRAILR2/TNFRH2, TNFRSF23-mDcTRAILR1/TNFRH1, and TNFRSF26-mTNFRH3) have been identified in mice only.16  The TNF receptor NGFR differs in its biologic activity from other members. Furthermore, the nomenclature of the TNF superfamily ligands has been standardized by an international commission as TNF superfamily (TNFSF), and the receptors as TNFRSF (Table 1). The cell types expressing each ligand or receptor are well characterized and summarized in Table 1. Both the ligands and receptors are expressed primarily by various cells of the immune system, but other cell types have been shown to express ligands as well as the receptors, under both physiologic and pathologic conditions. The importance of these proteins/cytokines in the pathophysiology of human diseases is indicated by the fact that in 2010 sales of TNF blockers exceeded US $20 billion (www.biospace.com/News).

Figure 2

Timeline for the discovery of various members of the TNF superfamily, their receptors, and the receptor-associated adaptor proteins.

Figure 2

Timeline for the discovery of various members of the TNF superfamily, their receptors, and the receptor-associated adaptor proteins.

Close modal
Table 1

Expression profile of ligands and receptors of human tumor necrosis factor superfamily

SymbolLigand (alias)Cellular expressionSymbolReceptor (alias)Cellular expression
TNFSF1 TNF-β (LT-α) NK, T, and B cells TNFRSF1A TNFR1 (DR1) Hematopoietic and immune cells 
   TNFRSF1B TNFR2 Immune and endothelial cells 
TNFSF2 TNF-α Macrophages and NK, T, and B cells TNFRSF1A/B TNFR1/2 Immune and endothelial cells 
TNFSF3 LT-β Activated CD4+ T cells and T, DC, and NK cells TNFRSF3 LT-βR NK cells, CD4+ and CD8+ T cells 
TNFSF4 OX40L (CD252, gp34) B and T cells, DCs, endothelial and smooth muscle cells TNFRSF4 OX40 (CD134) Activated CD4+ T cells and neutrophils 
TNFSF5 CD40L (CD154, gp39) Activated CD4+ T lymphocytes, NK cells, mast cells, basophils, and eosinophils TNFRSF5 CD40 (p50) B cells, monocytes, DCs, and thymic epithelium, Reed-Sternberg cells 
TNFSF6 FasL (CD95L, Apo1L) Activated splenocytes, thymocytes, nonlymphoid tissues, and NK cells TNFRSF6 Fas (CD95, Apo1, DR2) Epithelial cells, hepatocytes, activated mature lymphocytes, and transformed cells 
   TNFRSF6B DcR3 Lung and colon cells 
TNFSF7 CD27L (CD70) NK, T, B, and mast cells, smooth muscle and thymic epithelial cells TNFRSF7 CD27 Hematopoietic progenitors, and CD4+ and CD8+T cells 
TNFSF8 CD30L (CD153) Activated T cells, B cells, and monocytes, granulocytes, and medullary thymic epithelial cells TNFRSF8 CD30 Reed-Sternberg cells 
TNFSF9 4–1BBL APCs (B cells, macrophages, and DCs), mast cells TNFRSF9 4–1BB (CD137, ILA) T, NK, and mast cells, and neutrophils 
TNFSF10 TRAIL (Apo2L) NK and T cells, DCs TNFRSF10A TRAILR1 (DR4, Apo2) Most normal and transformed cells 
   TNFRSF10B TRAILR2 (DR5) Most normal and transformed cells 
   TNFRSF10C TRAILR3 (DcR1) Most normal and transformed cells 
   TNFRSF10D TRAILR4 (DcR2) Most normal and transformed cells 
   TNFRSF11B OPG (OCIF) Most normal and transformed cells 
TNFSF11 RANKL (TRANCE, OPGL, ODF) T cells, thymus, and lymph nodes TNFRSF11A RANK (TRANCER) Osteoclasts, osteoblasts, and activated T cells 
   TNFRSF11B OPG (OCIF) Osteoclast precursors, endothelial cells, and others 
TNFSF12 TWEAK (Apo3L) Monocytes TNFRSF12A TWEAKR (FN14) Endothelial cells and fibroblasts 
TNFSF13 APRIL (TALL-2, TRDL-1) Macrophages, lymphoid cells, and tumor cells TNFRSF13A/17 BCMA B cells, PBLs, spleen, thymus, lymph nodes, liver, and adrenals 
   TNFRSF13B TACI B cells, activated T cells, PBLs, spleen, thymus, and small intestine 
TNFSF13B BAFF (BLYS, THANK) T cells, monocytes, macrophages, and DCs TNFRSF13B TACI B cells, activated T cells, PBLs, spleen, thymus, and small intestine 
   TNFRSF13C BAFFR B cells, resting T cells, PBLs, spleen, lymph nodes 
   TNFRSF17 BCMA B cells, resting T cells, PBLs, spleen, lymph nodes 
TNFSF14 LIGHT (HVEML, LT-γ) T cells, granulocytes, monocytes, and DCs TNFRSF14 LIGHTR (HVEM) T and B cells, monocytes, and lymphoid cells 
   TNFRSF3 LT-βR Nonlymphoid hematopoietic and stromal cells 
TNFSF15 VEGI (TL1A) Endothelial cells TNFRSF25 DR3 NK cells, CD4+ and CD8+ T cells 
  APCs (B cells, macrophages, and DCs) TNFRSF6B DcR3 Activated T cells 
TNFSF18 GITRL HUVECs TNFRSF18 GITR (AITR) CD4+CD25+ T cells 
 EDA-A1 Skin  EDAR Ectodermal derivative 
 EDA-A2 Skin  XEDAR Ectodermal derivative, embryonic hair follicles 
NI   TNFRSF19 TROY (TAJ) Embryo skin, epithelium, hair follicles, and brain 
NI   TNFRSF19L RELT Lymphoid tissues, hematopoietic tissues 
NI   TNFRSF21 DR6 Resting T cells 
NI   TNFRSF16 NGFR (CD271) Neuronal axons, Schwann cells, perineural cells 
SymbolLigand (alias)Cellular expressionSymbolReceptor (alias)Cellular expression
TNFSF1 TNF-β (LT-α) NK, T, and B cells TNFRSF1A TNFR1 (DR1) Hematopoietic and immune cells 
   TNFRSF1B TNFR2 Immune and endothelial cells 
TNFSF2 TNF-α Macrophages and NK, T, and B cells TNFRSF1A/B TNFR1/2 Immune and endothelial cells 
TNFSF3 LT-β Activated CD4+ T cells and T, DC, and NK cells TNFRSF3 LT-βR NK cells, CD4+ and CD8+ T cells 
TNFSF4 OX40L (CD252, gp34) B and T cells, DCs, endothelial and smooth muscle cells TNFRSF4 OX40 (CD134) Activated CD4+ T cells and neutrophils 
TNFSF5 CD40L (CD154, gp39) Activated CD4+ T lymphocytes, NK cells, mast cells, basophils, and eosinophils TNFRSF5 CD40 (p50) B cells, monocytes, DCs, and thymic epithelium, Reed-Sternberg cells 
TNFSF6 FasL (CD95L, Apo1L) Activated splenocytes, thymocytes, nonlymphoid tissues, and NK cells TNFRSF6 Fas (CD95, Apo1, DR2) Epithelial cells, hepatocytes, activated mature lymphocytes, and transformed cells 
   TNFRSF6B DcR3 Lung and colon cells 
TNFSF7 CD27L (CD70) NK, T, B, and mast cells, smooth muscle and thymic epithelial cells TNFRSF7 CD27 Hematopoietic progenitors, and CD4+ and CD8+T cells 
TNFSF8 CD30L (CD153) Activated T cells, B cells, and monocytes, granulocytes, and medullary thymic epithelial cells TNFRSF8 CD30 Reed-Sternberg cells 
TNFSF9 4–1BBL APCs (B cells, macrophages, and DCs), mast cells TNFRSF9 4–1BB (CD137, ILA) T, NK, and mast cells, and neutrophils 
TNFSF10 TRAIL (Apo2L) NK and T cells, DCs TNFRSF10A TRAILR1 (DR4, Apo2) Most normal and transformed cells 
   TNFRSF10B TRAILR2 (DR5) Most normal and transformed cells 
   TNFRSF10C TRAILR3 (DcR1) Most normal and transformed cells 
   TNFRSF10D TRAILR4 (DcR2) Most normal and transformed cells 
   TNFRSF11B OPG (OCIF) Most normal and transformed cells 
TNFSF11 RANKL (TRANCE, OPGL, ODF) T cells, thymus, and lymph nodes TNFRSF11A RANK (TRANCER) Osteoclasts, osteoblasts, and activated T cells 
   TNFRSF11B OPG (OCIF) Osteoclast precursors, endothelial cells, and others 
TNFSF12 TWEAK (Apo3L) Monocytes TNFRSF12A TWEAKR (FN14) Endothelial cells and fibroblasts 
TNFSF13 APRIL (TALL-2, TRDL-1) Macrophages, lymphoid cells, and tumor cells TNFRSF13A/17 BCMA B cells, PBLs, spleen, thymus, lymph nodes, liver, and adrenals 
   TNFRSF13B TACI B cells, activated T cells, PBLs, spleen, thymus, and small intestine 
TNFSF13B BAFF (BLYS, THANK) T cells, monocytes, macrophages, and DCs TNFRSF13B TACI B cells, activated T cells, PBLs, spleen, thymus, and small intestine 
   TNFRSF13C BAFFR B cells, resting T cells, PBLs, spleen, lymph nodes 
   TNFRSF17 BCMA B cells, resting T cells, PBLs, spleen, lymph nodes 
TNFSF14 LIGHT (HVEML, LT-γ) T cells, granulocytes, monocytes, and DCs TNFRSF14 LIGHTR (HVEM) T and B cells, monocytes, and lymphoid cells 
   TNFRSF3 LT-βR Nonlymphoid hematopoietic and stromal cells 
TNFSF15 VEGI (TL1A) Endothelial cells TNFRSF25 DR3 NK cells, CD4+ and CD8+ T cells 
  APCs (B cells, macrophages, and DCs) TNFRSF6B DcR3 Activated T cells 
TNFSF18 GITRL HUVECs TNFRSF18 GITR (AITR) CD4+CD25+ T cells 
 EDA-A1 Skin  EDAR Ectodermal derivative 
 EDA-A2 Skin  XEDAR Ectodermal derivative, embryonic hair follicles 
NI   TNFRSF19 TROY (TAJ) Embryo skin, epithelium, hair follicles, and brain 
NI   TNFRSF19L RELT Lymphoid tissues, hematopoietic tissues 
NI   TNFRSF21 DR6 Resting T cells 
NI   TNFRSF16 NGFR (CD271) Neuronal axons, Schwann cells, perineural cells 

NI indicates not identified; OX40L, OX40 ligand; Fas, fibroblast-associated; TRANCE, TNF-related activation-induced cytokines; OPGL, OPG ligand; ODF, osteoclast differentiation factor; TALL, TNF- and APOL-related leukocyte expressed ligand; TRDL, TNF-related death ligand; HVEM, herpesvirus entry mediator; GITR, glucocorticoid-induced TND receptor; APCs, antigen-presenting cells; HUVECs, human umbilical vein endothelial cells; ILA, induced by lymphocyte activation; OCIF, osteoclastogenesis inhibitory factor; FN14, fibroblast growth factor-inducible immediate-early response gene 14; PBLs, peripheral blood lymphocytes; AITR, activation-inducible TNF receptor superfamily member; XEDAR, X-linked ectodysplasin receptor; TROY, TNFRSF expressed on the mouse embryo; TAJ, toxicity and JNK inducer; RELT, receptor expressed in lymphoid tissues; and NGFR, nerve growth factor receptor.

TNF-α was first identified as a factor with antitumor activity. The potent pro-inflammatory activity, however, prevents systemic administration of TNF-α to cancer patients.17,18  However, TNF-α is indeed used in the clinic. Specifically, it is used in the treatment of soft tissue sarcomas and melanomas in the extremities, using a technique called isolated limb perfusion. In this setting, TNF-α has demonstrated potent antitumor activity with an acceptable safety profile.19-21 

In the sections to follow, we discuss the signaling events mediated by members of TNF superfamily and receptors with a particular emphasis on TNF-α. We describe the role of this superfamily in normal physiology and disease. Finally, we discuss rationally designed therapeutics that are based on TNF signaling.

The mechanism by which TNF and its various family members transduce target cell signals has been studied extensively. That the 19 members of the TNF superfamily interact with 29 distinct receptors implies that at least some of the ligands must interact with more than one receptor. For instance, TNF-α is known to interact with 2 distinct receptors, TNF receptor 1 (TNFR1) and TNFR2 (Figure 3). On B cells, BAFF has been shown to bind to 3 distinct receptors, transmembrane activator and CAML interactor (TACI), B-cell maturation protein (BCMA), and BAFF receptor (BAFFR). TRAIL has been shown to bind to as many as 5 different receptors, including death receptor 4 (DR4), DR5, decoy receptor 1 (DcR1), DcR2, and osteoprotegerin (OPG).22  Whereas DR4 and DR5 transduce the signals across the cell membrane, DcR1 and DcR2 bind the ligand but do not transduce signals and OPG is primarily a soluble receptor without any transmembrane domain. Some of the cell-surface receptors bind more than one ligand, as in the case of TNF-α and TNF-β, both of which bind to the same receptor with comparable affinity.8 

Figure 3

Autocrine, paracrine, cell to cell, and reverse signaling pathways for TNF-α. Ligands, such as TNF-α, are expressed as both transmembrane and in soluble forms. The transmembrane form of the ligand appears to mediate therapeutic effects, but soluble ligand is linked to pathologic effects of TNF-α. Ligands, such as TNF-β, lack the transmembrane domain and thus are expressed only as a soluble protein. TNF-α, made by tumor cells, acts primarily through TNFR1 in an autocrine and paracrine manner. There are also examples of reverse signaling through TNF-α when it binds to its receptor.

Figure 3

Autocrine, paracrine, cell to cell, and reverse signaling pathways for TNF-α. Ligands, such as TNF-α, are expressed as both transmembrane and in soluble forms. The transmembrane form of the ligand appears to mediate therapeutic effects, but soluble ligand is linked to pathologic effects of TNF-α. Ligands, such as TNF-β, lack the transmembrane domain and thus are expressed only as a soluble protein. TNF-α, made by tumor cells, acts primarily through TNFR1 in an autocrine and paracrine manner. There are also examples of reverse signaling through TNF-α when it binds to its receptor.

Close modal

Most of the receptors of the TNF superfamily can be classified into one of 2 categories: those that possess an intracellular death domain (DD) and those that do not. The DD is a region approximately 45 amino acids long that is required for recruitment of other proteins resulting in cell death. Six different receptors have been identified with DD in their intracellular domain: DR1 (also called TNFR1); DR2 (also called Fas); DR3, to which VEGI binds; DR4 and DR5, to which TRAIL binds; and DR6. No ligand has yet been identified that binds to DR6. Interestingly, a recent study indicates that DR6 is broadly expressed by developing neurons and is activated by β-amyloid precursor protein.23  The authors suggested that an extracellular fragment of β-amyloid precursor protein, acting via DR6 and caspase-6, contributes to Alzheimer disease.

The expression of various receptors of the TNF superfamily may vary significantly between cell types and tissue. Receptors that contain a DD in their intracellular domain are expressed most universally. TNF receptor 1 (TNFR1), which contains a DD, is highly promiscuous and is expressed on every cell type in the body studied to date. This may reflect the receptor's diverse functions in different cell types. Similarly, DR4 and DR5 have been shown to be expressed in most cell types. The expression of TNFR2, in comparison, is limited to cells of the immune system, endothelial cells, and nerve cells. Fas receptor expression is also highly heterogeneous and is found on epithelial cells, hepatocytes, and lymphocytes.

Cell signaling for most cytokines and growth factors is normally mediated through interaction between a soluble ligand and a transmembrane receptor. Among the TNF superfamily, however, several ligands have been identified that rarely appear as soluble ligand, including FasL, CD27L, CD30L, CD40L, OX40L, and 4-1BBL. All of these ligands are instead primarily expressed as transmembrane proteins on the cell surface and interact with cells that express the corresponding receptors. Some ligands, such as TNF-α, are expressed as both transmembrane and soluble forms, whereas ligands, such as TNF-β, lack the transmembrane domain and thus are expressed only as a soluble protein. Obviously, cell signaling effects are more restricted when a ligand is ex-pressed only as a transmembrane protein. There are reports, furthermore, that the nature of cell signaling may differ between ligands expressed as soluble protein and those expressed in transmembrane form.3 

Another characteristic feature of some ligand-receptor interactions in the TNF superfamily is the phenomenon of “reverse signaling”: instead of signaling being transmitted from ligand to receptor, it is transmitted from receptor to the cell bearing the transmembrane form of the ligand (Figure 3).24  For example, one report showed that TNF-α, when activated by TNF-α antibody, induced E-selectin (CD62E) expression on activated human CD4+ T cells via an outside-to-inside signal through the membrane.25  This report indicates that membrane TNF-α can transmit bipolar signals. Reverse signaling has also been reported via the CD30 ligand.26  CD30 ligand is a type II membrane protein with a C-terminal extracellular domain that is homologous with the extracellular domains of other TNF family members. Cross-linking of CD30 ligand by a monoclonal antibody or by CD30-Fc fusion protein induced production of IL-8 by freshly isolated neutrophils. CD30 ligand, but not CD30, is expressed on neutrophils. Clearly, several TNF family members and their cognate receptors signal bidirectionally, thus blurring the distinction between ligand and receptor. How this kind of signaling is mediated is not fully understood.

TNF-α induces at least 5 different types of signals that include activation of NF-κB, apoptosis pathways, extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38MAPK), and c-Jun N-terminal kinase (JNK; Figure 4). When TNF-α binds to TNFR1, it recruits a protein called TNFR-associated death domain (TRADD) through its DD.27  TRADD then recruits a protein called Fas-associated protein with death domain (FADD), which then sequentially activates caspase-8 and caspase-3, and thus apoptosis.28  Alternatively, TNF-α can activate mitochondria to sequentially release ROS, cytochrome C, and Bax, leading to activation of caspase-9 and caspase-3 and thus apoptosis.29  Paradoxically, TNF-α has also been shown to activate NF-κB, which in turn regulates the expression of proteins associated with cell survival and cell proliferation.30  NF-κB activation by TNF-α is mediated through sequential recruitment of TNFR1, TRADD, TNFR-associated factor 2 (TRAF2/TRAF5), receptor interacting protein (RIP), TGF-β–activated kinase 1 (TAK1), IκB kinase (IKK) complex, and inhibitor of nuclear factor-κBα (IκBα) phosphorylation, ubiquitination, and degradation, and finally nuclear translocation of p50 and p65 and DNA binding.31  The pro-inflammatory effect of TNF is mediated through NF-κB–regulated proteins, such as IL-6, IL-8, IL-18, chemokines, inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and 5-lipoxygenase (5-LOX), all major mediators of inflammation. Indeed, TNF-α can induce expression of TNF-α itself through activation of NF-κB.3  TNF-α can also activate cellular proliferation through activation of another transcription factor, activator protein-1 (AP-1),32  which is activated by TNF-α through sequential recruitment of TNFR1, TRADD, TRAF2, MAP/ERK kinase kinase 1 (MEKK1), MAP kinase kinase 7 (MKK7), and JNK. The activation of p38MAPK by TNF-α is mediated through TRADD-TRAF2-MKK3. How TNFR2, which lacks a DD, activates cell signaling is much less clear than how TNFR1 activates cell signaling. Because TNFR2 can directly bind to TRAF2, it can activate both NF-κB and MAPK signaling quite well. Interestingly, TRADD has been reported recently to mediate cell signaling by TOLL-like receptors 3 and 4.33 

Figure 4

Cell signaling pathways activated by TNF. TNFR1 activation leads to recruitment of intracellular adaptor proteins (TRADD, FADD, TRAF, and RIP), which activate multiple signal transduction pathways. TNFR sequentially recruits TRADD, TRAF2, RIP, TAK1, and IKK, leading to the activation of NF-κB31 ; and the recruitment of TRADD, FADD, and caspase-8, leads to the activation of caspase-3, which in turn induces apoptosis.28  JNK is activated through the sequential recruitment of TRAF2, RIP, MEKK1, and MKK7.79  Exposure of cells to TNFα in most cases results in the generation of reactive oxygen species, leading to activation of MKK7 and JNK.29  The activation of ERK and p38MAPK is via TRADD, TRAF2, RIP, TAK1, and MKK3/6.80 

Figure 4

Cell signaling pathways activated by TNF. TNFR1 activation leads to recruitment of intracellular adaptor proteins (TRADD, FADD, TRAF, and RIP), which activate multiple signal transduction pathways. TNFR sequentially recruits TRADD, TRAF2, RIP, TAK1, and IKK, leading to the activation of NF-κB31 ; and the recruitment of TRADD, FADD, and caspase-8, leads to the activation of caspase-3, which in turn induces apoptosis.28  JNK is activated through the sequential recruitment of TRAF2, RIP, MEKK1, and MKK7.79  Exposure of cells to TNFα in most cases results in the generation of reactive oxygen species, leading to activation of MKK7 and JNK.29  The activation of ERK and p38MAPK is via TRADD, TRAF2, RIP, TAK1, and MKK3/6.80 

Close modal

Although members of the TNF superfamily have the potential to activate NF-κB, almost all of them, unlike TNF-α, activate NF-κB in a cell type-specific manner. This is restricted in part by the expression of cell surface receptors on particular cell types and in part through independent mechanisms yet to be determined. For instance, DR2, DR4, and DR5 are expressed on most cell types, but their interactions with respective ligands rarely lead to NF-κB activation. On the other hand, interaction of DR1 with its ligand leads to NF-κB activation in most cells. This ability of TNF-α to activate NF-κB nonselectively makes it the most promiscuous and universal pro-inflammatory family member in most situations. TNF-β has been shown to be a less potent pro-inflammatory agent than TNF-α.34 

Although various members of the TNF superfamily are known to activate apoptosis, most cells are resistant to apoptosis induced by TNF-α alone.35  The mechanism for this resistance to apoptosis is not understood; it is not the result of lack of receptors. It seems that cell survival, proliferative, and apoptotic signals are all activated simultaneously by TNF-α and related proteins and that the balance between these signals determines whether the TNF family member induces apoptosis, proliferation, versus no effect at all. Whereas new protein synthesis is usually required for survival and proliferative signals, no protein synthesis is required for apoptosis.

Although initially discovered as an anticancer agent, TNF-α and its family members have now been linked to an array of pathophysiologies, including cancer, neurologic diseases, cardiovascular diseases, pulmonary diseases, autoimmune diseases, and metabolic diseases.

TNF-α and cancer

The carcinogenic activities of TNF-α are mediated through its ability to activate the pro-inflammatory transcription factor NF-κB, which up-regulates the expression of genes linked to tumor cell survival, proliferation, invasion, angiogenesis, and metastasis.36  Several tumor cell types constitutively express TNF-α, including ovarian cancer, breast cancer, and others.37  Most tumor cells that express TNF-α exhibit constitutive activation of NF-κB. These tumors cells are “addicted” to NF-κB, as their survival is highly dependent on this factor.

In contrast to TNF-α, other members of the TNF superfamily exhibit more anticancer as opposed to oncogenic potential. For instance, TRAIL is being actively explored as an anticancer agent because of its preferential ability to induce apoptosis in tumorigenic or transformed cells, but not in normal cells or tissues.38  Like TNF-α, CD27L, CD30L, CD40L, APRIL, and BAFF, although essential regulators of the immune system, play a major role in hematopoietic tumorigenesis.

TNF-α and neurologic diseases

Both TNF-α and its receptors are expressed by microglial cells in the brain. Through activation of NF-κB, TNF-α plays an essential role in the survival of these cells. In the brain, TNF-α has been shown to induce pro-inflammatory signals that have been linked with depression,39  bipolar disporder,40  epilepsy,41  Alzheimer disease,42  Parkinson disease,43  and multiple sclerosis.44  Similarly, Fas ligation has been shown to induce selective expression of chemokines, IL-8, and monocyte chemoattractant protein-1 (MCP-1) in human astroglioma cells in vitro.45 

TNF-α and cardiovascular diseases

TNF-α, along with other inflammatory molecules, is known to play a role in the initiation and progression of cardiovascular diseases. Although normal heart does not express TNF-α, the failing heart produces massive amounts of TNF-α. There is now growing evidence that the immune system is an important source for TNF production in failing heart. However, myocardium may also synthesize TNF-α de novo in failing heart.46  Perhaps the earliest indication that TNF-α is linked with heart failure emerged in 1990 when levels of circulating TNF-α levels were found to be elevated in sera of patients with chronic heart failure.47  Since then, numerous reports and reviews have been published on this subject.48  Evidence indicate that sustained and excessive production of TNF-α worsens the disease.49 

Reports indicate that patients with chronic inflammatory disorders, such as rheumatoid arthritis (RA) and psoriasis, exhibit higher than expected rates of mortality that cannot be explained by traditional risk factors alone.50  This statement implies that inflammatory pathways might also contribute to the increased vascular risk in such diseases. The epidemiologic, physiologic, and model data suggest that TNF-α is directly involved in vascular pathophysiology. According to one published report, there are approximately 50% greater chances of cardiovascular mortality in RA patients compared with the general population.51 

TNF-α and pulmonary diseases

TNF-α has been shown to play a major role in various pulmonary diseases, including asthma, chronic bronchitis, chronic obstructive pulmonary disease, acute lung injury, and acute respiratory distress syndrome.52  TNF-α is expressed in asthmatic airways and has been shown to play a role in amplifying asthmatic inflammation through the activation of NF-κB, AP-1, and other transcription factors.53  TNF-α has also been shown to induce pathologic features associated with COPD in animal models, such as an inflammatory cell infiltrate into the lungs, pulmonary fibrosis, and emphysema.54  TRAIL has also been linked to an inflammatory response in asthma.55  Interestingly, although TNF-α and TRAIL mediate lung inflammation, signaling through DR3 has been linked with expansion of Tregs, which plays a protective role against allergic lung inflammation in a mouse model of asthma.56 

TNF-α, diabetes, and obesity

The first indication of increased cytokine release in obesity was provided by the identification of increased expression of TNF-α in the adipose tissue of obese mice in the early 1990s. TNF-α is expressed in and secreted by adipose tissue; its levels correlate with the degree of adiposity and the associated insulin resistance.57  The dephosphorylation of insulin receptor substrate-1 through activation of protein phosphatases has been shown to account for TNF-α-induced insulin resistance.57  Obesity is a leading cause of insulin resistance and type 2 diabetes, and targeting TNF-α and/or its receptors has been suggested as a promising treatment strategy for these conditions.58  Emerging evidence has indicated that inflammation is one of the critical processes associated with development of insulin resistance, diabetes, and related diseases, and obesity is now considered a state of chronic low-grade inflammation.59 

TNF-α and autoimmune diseases

One area where TNF family members play a pivotal role is in autoimmune diseases. Uveitis, multiple sclerosis, systemic lupus, arthritis, psoriasis, and Crohn disease are all autoimmune diseases and are connected with dysregulation of various members of the TNF superfamily or their receptors as described here.60  Indeed, TNF-α blockers have been approved for the treatment of osteoarthritis, psoriasis, and Crohn disease. Among all the members of the TNF superfamily, the role of TNF-α in autoimmune diseases is best understood.

From the studies already described, it is clear that both TNF-α and its family members play critical roles in the immune, cardiovascular, pulmonary, metabolic, and neurologic systems. Further insights into the function of these cytokines have been gained through gene deletion/gene knockout studies. The genes for the ligands and the receptors of the TNF superfamily have been deleted one by one in mice, and the resulting phenotypes have been examined (Table 2). Most commonly, defects in the immune system have been observed in these mice. For instance, deletion of TNF-β yielded a defect in development of secondary lymphoid organs and disorganized splenic architecture.61  Deletion of TNF-α, on the other hand, resulted in normal development and a complete lack of detectable morphologic or gross structural abnormalities.62  However, TNF-α-deficient mice were highly susceptible to challenge with an infectious agent and were resistant to the lethality of minute doses of lipopolysaccharide after D-galactosamine treatment.63  Amazingly, deletion of TNF-α had no effect on phagocytic activity of macrophages or on T-cell functions, as measured by proliferation, cytokine release, or cytotoxicity, suggesting redundancy with other superfamily members.

Table 2

Effect of gene knockout on the phenotype of TNF superfamily, receptors, and receptor-associated proteins

GenePhenotype
Cytokine  
    TNF-β (LT-α) Defects in secondary lymphoid organ development; disorganized splenic microarchitecture61  
    TNF-α No phenotypic abnormalities in LN; lack splenic primary B-cell follicles; disorganized FDC networks and germinal centers62  
    LT-β Defects in organogenesis of the lymphoid system; lymphocytosis in the circulation and peritoneal cavity; lymphocytic infiltrations in lungs and liver81  
    OX40L Defective T-cell responses82  
    CD40L Defective T-cell and IgG responses; hyper-IgM syndrome82  
    FasL Impaired activation-induced T-cell death; lymphoproliferation; autoimmunity82  
    4–1BBL Defective T-cell responses82  
    TRAIL Delayed regression of retinal neovascularization64  
    RANKL Osteopetrosis; growth retardation of limbs, skull, and vertebrae; chondrodysplasia83  
    APRIL Normal immune system development 84  
 Impaired IgA class switching85  
    BAFF Impaired B-cell maturation86  
 Low Ig serum levels; block in B-cell development at the T1 stage; absence of T2, mantle, and follicular zone B cells in the LN and spleen87  
    EDA-A1 Ectodermal dysplasias88 * 
    EDA-A2 Impaired development of hair, eccrine sweat glands, and teeth89  
 Multifocal myodegeneration90  
Receptor  
    TNFR1 Resistant to low levels of LPS; increased susceptibility to Listeria monocytogenes infection91  
 Impaired oval cell proliferation; reduction of tumorigenesis92  
    TNFR2 Increased sensitivity to bacterial pathogens; decreased sensitivity to LPS; reduced antigen-induced T-cell 
 apoptosis82  
    LT-βR Absence of LN, PP; defective GC formation82  
    OX40 Defective T-cell responses82  
    CD40 Defective Ig class switching and GC formation causing immunodeficiency82  
    Fas Impaired activation-induced T-cell death; lymphoproliferative syndrome; autoimmunity93  
 Accumulation of autoreactive B cells in T cell-rich zones; production of autoantibodies94  
 Resistant to suppression by high doses of antigen and to apoptosis in mature CD4+ T cells95  
    CD27 Defective T-cell responses82  
    CD30 Impaired follicular GC responses; reduced recall-memory Ab responses96  
    4–1BB Enhanced T-cell response but normal T-cell development97  
 Reduced number of NK and NKT cells; resistance to LPS-induced shock syndrome98  
 Increased number of myeloid progenitor and mature DCs; impaired DC function99  
 Reduced atherosclerosis in hyperlipidemic mice100  
    DR5 Normal development with an enlarged thymus66  
    RANK Osteopetrosis; absence of osteoclasts and LN; PP present; abnormal B-cell development82  
    OPG Osteoporosis; arterial calcification82  
    FN14 Reduced proliferative capacity; altered myotube formation101  
 Reduced neurogenesis in the subventricular zone102  
 Reduced LPC numbers; attenuated inflammation; cytokine production103  
    TACI Increased B-cell accumulation; splenomegaly104  
    BAFFR Reduced late transitional and follicular B-cell numbers; devoid of marginal zone B cells; reduced CD21 and CD23 surface expression105  
    DR3 Impaired negative selection and anti–CD3-induced apoptosis106  
    GITR Abolished anti–CD3-induced T-cell activation107  
    EDAR Abnormal tooth, hair, and sweat gland formation82  
    XEDAR No different than wild-type littermates90  
    TROY No apparent defects in skin appendages108  
    DR6 Enhanced CD4+ T-cell expansion and Th2 differentiation; enhanced splenic GC formation109  
 Impaired JNK activity; T-cell differentiation110  
 CD4+ T-cell proliferation; Th differentiation111  
    NGFR Decreased sensory neuron innervation; impaired heat sensitivity112  
Receptor-associated proteins  
    TRAF1 Normal lymphocyte development113  
 Attenuation of atherosclerosis114  
    TRAF2 Died prematurely; elevated sTNF levels; hypersensitivity to TNF-induced cell death115  
    TRAF3 Postnatal lethality; defect in T-dependent immune responses116  
    TRAF5 Defect in proliferation; up-regulation of surface molecules CD23, CD54, CD80, CD86, and Fas after CD40 stimulation117  
    TRAF6 Osteopetrosis; perinatal and postnatal lethality118  
 Hypohidrotic ectodermal dysplasia119  
    TRADD Embryonic lethal120  
    FADD Embryonic lethal; cardiac failure and abdominal hemorrhage121  
    FAN Impaired neutral sphingomyelinase activation; cutaneous barrier repair122  
    FLICE Perinatal lethal123  
    RIP Perinatal lethal124  
    IKK-α Abnormal morphogenesis (limb and skeletal patterning; proliferation and differentiation of epidermal keratinocytes)125  
    IKK-β Died at mid-gestation126  
    Apaf-1 Reduced apoptosis in the brain; hyperproliferation of neuronal cells127  
    Caspase-9 Embryonic lethal; defect in brain development128  
    Caspase-3 Decreased apoptosis in the brain; premature lethality129  
    EDARADD Hypohidrotic ectodermal dysplasia130  
    Act1 B cell–mediated autoimmune phenotypes131  
GenePhenotype
Cytokine  
    TNF-β (LT-α) Defects in secondary lymphoid organ development; disorganized splenic microarchitecture61  
    TNF-α No phenotypic abnormalities in LN; lack splenic primary B-cell follicles; disorganized FDC networks and germinal centers62  
    LT-β Defects in organogenesis of the lymphoid system; lymphocytosis in the circulation and peritoneal cavity; lymphocytic infiltrations in lungs and liver81  
    OX40L Defective T-cell responses82  
    CD40L Defective T-cell and IgG responses; hyper-IgM syndrome82  
    FasL Impaired activation-induced T-cell death; lymphoproliferation; autoimmunity82  
    4–1BBL Defective T-cell responses82  
    TRAIL Delayed regression of retinal neovascularization64  
    RANKL Osteopetrosis; growth retardation of limbs, skull, and vertebrae; chondrodysplasia83  
    APRIL Normal immune system development 84  
 Impaired IgA class switching85  
    BAFF Impaired B-cell maturation86  
 Low Ig serum levels; block in B-cell development at the T1 stage; absence of T2, mantle, and follicular zone B cells in the LN and spleen87  
    EDA-A1 Ectodermal dysplasias88 * 
    EDA-A2 Impaired development of hair, eccrine sweat glands, and teeth89  
 Multifocal myodegeneration90  
Receptor  
    TNFR1 Resistant to low levels of LPS; increased susceptibility to Listeria monocytogenes infection91  
 Impaired oval cell proliferation; reduction of tumorigenesis92  
    TNFR2 Increased sensitivity to bacterial pathogens; decreased sensitivity to LPS; reduced antigen-induced T-cell 
 apoptosis82  
    LT-βR Absence of LN, PP; defective GC formation82  
    OX40 Defective T-cell responses82  
    CD40 Defective Ig class switching and GC formation causing immunodeficiency82  
    Fas Impaired activation-induced T-cell death; lymphoproliferative syndrome; autoimmunity93  
 Accumulation of autoreactive B cells in T cell-rich zones; production of autoantibodies94  
 Resistant to suppression by high doses of antigen and to apoptosis in mature CD4+ T cells95  
    CD27 Defective T-cell responses82  
    CD30 Impaired follicular GC responses; reduced recall-memory Ab responses96  
    4–1BB Enhanced T-cell response but normal T-cell development97  
 Reduced number of NK and NKT cells; resistance to LPS-induced shock syndrome98  
 Increased number of myeloid progenitor and mature DCs; impaired DC function99  
 Reduced atherosclerosis in hyperlipidemic mice100  
    DR5 Normal development with an enlarged thymus66  
    RANK Osteopetrosis; absence of osteoclasts and LN; PP present; abnormal B-cell development82  
    OPG Osteoporosis; arterial calcification82  
    FN14 Reduced proliferative capacity; altered myotube formation101  
 Reduced neurogenesis in the subventricular zone102  
 Reduced LPC numbers; attenuated inflammation; cytokine production103  
    TACI Increased B-cell accumulation; splenomegaly104  
    BAFFR Reduced late transitional and follicular B-cell numbers; devoid of marginal zone B cells; reduced CD21 and CD23 surface expression105  
    DR3 Impaired negative selection and anti–CD3-induced apoptosis106  
    GITR Abolished anti–CD3-induced T-cell activation107  
    EDAR Abnormal tooth, hair, and sweat gland formation82  
    XEDAR No different than wild-type littermates90  
    TROY No apparent defects in skin appendages108  
    DR6 Enhanced CD4+ T-cell expansion and Th2 differentiation; enhanced splenic GC formation109  
 Impaired JNK activity; T-cell differentiation110  
 CD4+ T-cell proliferation; Th differentiation111  
    NGFR Decreased sensory neuron innervation; impaired heat sensitivity112  
Receptor-associated proteins  
    TRAF1 Normal lymphocyte development113  
 Attenuation of atherosclerosis114  
    TRAF2 Died prematurely; elevated sTNF levels; hypersensitivity to TNF-induced cell death115  
    TRAF3 Postnatal lethality; defect in T-dependent immune responses116  
    TRAF5 Defect in proliferation; up-regulation of surface molecules CD23, CD54, CD80, CD86, and Fas after CD40 stimulation117  
    TRAF6 Osteopetrosis; perinatal and postnatal lethality118  
 Hypohidrotic ectodermal dysplasia119  
    TRADD Embryonic lethal120  
    FADD Embryonic lethal; cardiac failure and abdominal hemorrhage121  
    FAN Impaired neutral sphingomyelinase activation; cutaneous barrier repair122  
    FLICE Perinatal lethal123  
    RIP Perinatal lethal124  
    IKK-α Abnormal morphogenesis (limb and skeletal patterning; proliferation and differentiation of epidermal keratinocytes)125  
    IKK-β Died at mid-gestation126  
    Apaf-1 Reduced apoptosis in the brain; hyperproliferation of neuronal cells127  
    Caspase-9 Embryonic lethal; defect in brain development128  
    Caspase-3 Decreased apoptosis in the brain; premature lethality129  
    EDARADD Hypohidrotic ectodermal dysplasia130  
    Act1 B cell–mediated autoimmune phenotypes131  

Fas indicates fibroblast-associated; FN14, fibroblast growth factor-inducible immediate-early response gene 14; GITR, glucocorticoid-induced TND receptor; EDAR, EDA receptor; XEDAR, X-linked ectodysplasin receptor; TROY, TNFRSF expressed on the mouse embryo; NGFR, nerve growth factor receptor; FAN, factor associated with neutral SMase activation; FLICE, FADD-like IL-1β-converting enzyme; Apaf-1, apoptotic protease activating factor-1; EDARADD, ectodysplasin-A receptor-associated adapter protein; LN, lymph node; FDC, follicular dendritic cell; LPS, lipopolysaccharide; PP, Peyer patches; GC, germinal center; and sTNF, serum TNF.

*

Study performed in human, others in mouse model.

The deletion of the TRAIL gene has been shown to affect the retinal vascularization in mice.64  Generally, deletions of a ligand and its receptor have been shown to produce related phenotypes; however, in some cases, the 2 phenotypes are quite dissimilar. For instance, TRAIL is known to bind 2 different signaling receptors, DR4 and DR5, in humans but only DR5 in mice. Whereas deletion of TRAIL affected susceptibility to tumor burden and accelerated autoimmune diseases,65  animals with deletion of DR5 developed normally but had an enlarged thymus.66  This indicates that the ligand may also function independently of the receptor.

Various mutations in genes for TNF, its family members, and its receptors have been identified in humans (Table 3). These mutations are reflected either by a change in phenotype or a change in function. For instance, abnormalities in the CD40L gene have been linked to the X-linked immunodeficiency hyper-IgM syndrome. This human disease is characterized by elevated concentrations of serum IgM and decreased amounts of all other immunoglobulin isotypes. The CD40L protein produced in these patients was incapable of binding to CD40 and thus was unable to induce proliferation or IgE secretion from normal B cells. Activated T cells from some of these affected patients failed to express functional CD40L.

Table 3

Diseases caused by mutation of TNF superfamily, receptors, and adaptors

GeneStudyDisease
Cytokine   
    TNF-β (LT-α) Human Cerebral infarction68  
    TNF-α Human Cerebral infarction68  
    CD40L Human X-linked hyper-IgM syndrome132  
    FasL Mouse Generalized lymphoproliferative disease133  
    EDA-A1 Human Ectodermal dysplasias88  
    EDA-A2 Dog X-linked hypohidrotic ectodermal dysplasia134  
Receptor   
    TNFR1 Human TNFR1-associated periodic syndrome135  
 Human TRAPS associated with SLE67  
 Human Crohn disease136  
    TNFR2 Human Crohn disease136  
    Fas Mouse Autoimmune lymphoproliferative syndrome137  
 Human Generalized lymphoproliferative disease133  
    RANK Human Familial expansile osteolysis138  
    OPG Human Idiopathic hyperphosphatasia139  
 Human Juvenile Paget disease140  
    TACI Human Common variable immunodeficiency141  
    BAFFR Mouse Lupus-like syndrome (B cell−mediated autoimmunity)142  
    EDAR Human Hypohidrotic ectodermal dysplasia143  
Adaptors   
    TRAF3 Human Herpes simplex encephalitis144  
    TRAF6 Mouse Hypohidrotic ectodermal dysplasia119  
    EDARADD Mouse Hypohidrotic ectodermal dysplasia130  
    Act1 Mouse B cell−mediated autoimmune phenotypes131  
GeneStudyDisease
Cytokine   
    TNF-β (LT-α) Human Cerebral infarction68  
    TNF-α Human Cerebral infarction68  
    CD40L Human X-linked hyper-IgM syndrome132  
    FasL Mouse Generalized lymphoproliferative disease133  
    EDA-A1 Human Ectodermal dysplasias88  
    EDA-A2 Dog X-linked hypohidrotic ectodermal dysplasia134  
Receptor   
    TNFR1 Human TNFR1-associated periodic syndrome135  
 Human TRAPS associated with SLE67  
 Human Crohn disease136  
    TNFR2 Human Crohn disease136  
    Fas Mouse Autoimmune lymphoproliferative syndrome137  
 Human Generalized lymphoproliferative disease133  
    RANK Human Familial expansile osteolysis138  
    OPG Human Idiopathic hyperphosphatasia139  
 Human Juvenile Paget disease140  
    TACI Human Common variable immunodeficiency141  
    BAFFR Mouse Lupus-like syndrome (B cell−mediated autoimmunity)142  
    EDAR Human Hypohidrotic ectodermal dysplasia143  
Adaptors   
    TRAF3 Human Herpes simplex encephalitis144  
    TRAF6 Mouse Hypohidrotic ectodermal dysplasia119  
    EDARADD Mouse Hypohidrotic ectodermal dysplasia130  
    Act1 Mouse B cell−mediated autoimmune phenotypes131  

RANK indicates receptor activator of NF-κB; OPG, osteoprotegerin; EDAR, EDA receptor; and EDARADD, ectodysplasin-A receptor-associated adapter protein.

TNFR1-associated periodic syndrome (TRAPS) is the result of a mutation in the TNF receptor gene and is associated with systemic lupus erythematosus (SLE).67  Two biallelic polymorphisms in TNF-α (TNF-α–308) and TNF-β (TNF-β +252) genes have been associated with TNF production and susceptibility to inflammatory diseases.68  Two genetic polymorphisms in the TNF locus (TNF-α–308 G → A and TNF-β +252 A → G) were found to be risk factors for cerebral infarction.68  These mutations provide novel insights into the function of various members of the TNF superfamily.

TNF superfamily members play a role in the pathogenesis of various human diseases (Figures 5 and 6). Thus, this superfamily represents an active target for drug development. Various antagonists against TNF family members, and their receptors have been approved by FDA and some are in clinical trials (Table 4). These agents are effective against RA, psoriatic arthritis, psoriasis, ulcerative colitis, and Crohn disease. Approved therapeutics are primarily one of 2 types of antagonist, either a soluble receptor that binds circulating TNF-α and acts as a “sink,” preventing interaction with cell surface receptors, or antibodies against TNF-α. Golimumab is a fully humanized TNF-α monoclonal antibody that is specific for human TNF-α and was approved for RA, psoriatic arthritis, and active ankylosing spondylitis.69  Similarly, Certolizumab pegol, a PEGylated Fab′ fragment of a humanized TNF antibody, is a monoclonal antibody directed against human TNF-α. Certolizumab pegol was approved for RA and Crohn disease.70  A human antibody against RANKL (denosumab, Prolia) was recently approved for the treatment of osteoporosis.

Figure 5

Various diseases that have been closely linked to TNF-α and members of its family.

Figure 5

Various diseases that have been closely linked to TNF-α and members of its family.

Close modal
Figure 6

Diseases caused by mutation of genes in members of the TNF superfamily, its receptors, and adaptor proteins. Red circle represents studies in human; and yellow circle, studies in mice. Asterisk (*) within yellow circle indicates diseases in dogs.

Figure 6

Diseases caused by mutation of genes in members of the TNF superfamily, its receptors, and adaptor proteins. Red circle represents studies in human; and yellow circle, studies in mice. Asterisk (*) within yellow circle indicates diseases in dogs.

Close modal
Table 4

Selected FDA-approved drugs and those in development targeting the TNF superfamily

CytokineDrugMechanismDiseaseStatus
TNF-α Infliximab mAbs RA, PA, psoriasis, ALS, ulcerative colitis, Approved 
   Crohn disease  
 Etanercept RD RA, PA, psoriasis, ALS, juvenile RA Approved 
 Adalimumab mAbs RA, PA, psoriasis, ALS, juvenile RA, Crohn disease Approved 
 Certolizumab mAbs RA, Crohn disease Approved 
 Golimumab mAbs RA, PA, AS Approved 
 TNF-α kinoid Vaccine* RA, Crohn disease Phase 2 
 ESBA105 SC antibody Anterior uveitis Phase 2 
 ART621 mAbs RA, psoriasis Phase 2 
 ATN-103 Nanobody RA Phase 1 
LT-β Baminercept α LT-βR-Ig RA Phase 2 
OX40L huMAb OX40L mAbs Asthma Phase 2 
CD40L Dacetuzumab mAbs Multiple myeloma Phase 1 
   Lymphoma, large B-cell, diffuse lymphoma, NHL Phase 2 
 HCD122 mAbs Multiple myeloma Phase 1 
   Follicular lymphoma Phase 2 
FasL APO010 Fusion protein Solid tumors Phase 1 
CD27L MDX-1411 mAbs Kidney cancer Phase 1 
 MDX-1203 ADC Renal cell carcinoma, NHL Phase 1 
CD30L XmAb2513 mAbs Hodgkin lymphoma, anaplastic large-cell lymphoma Phase 1 
 SGN-35 ADC Hodgkin lymphomas Phase 3 
   Lymphoma, large-cell, anaplastic lymphoma, NHL Phase 2 
 MDX-1401 mAbs Hodgkin lymphomas Phase 1 
4–1BBL BMS-663513 mAbs Advanced cancer Phase 1 
   Melanoma Phase 2 
TRAIL Mapatumumab mAbs Colorectal, NHL, non−small-cell lung cancer Phase 2 
 Lexatumumab mAbs Kidney cancer, lymphoma, neuroblastoma, sarcoma Phase 1 
 CS-1008 mAbs Malignancies, lymphoma Phase 1 
   Colorectal neoplasms, pancreatic, lung cancer Phase 2 
RANKL Denosumab mAbs Low bone mass, low bone mineral density, osteopenia, osteoporosis Phase 3 
TWEAK BIIB023 mAbs RA Phase 1 
APRIL Atacicept Fusion protein RA Phase 2 
   SLE Phase 3 
BAFF Atacicept Fusion protein RA Phase 2 
 Benlysta mAbs Arthritis, rheumatoid, SLE Phase 2 
 LY2127399 mAbs RA Phase 2 
LIGHT Baminercept α LT-βR-Ig RA Phase 2 
CytokineDrugMechanismDiseaseStatus
TNF-α Infliximab mAbs RA, PA, psoriasis, ALS, ulcerative colitis, Approved 
   Crohn disease  
 Etanercept RD RA, PA, psoriasis, ALS, juvenile RA Approved 
 Adalimumab mAbs RA, PA, psoriasis, ALS, juvenile RA, Crohn disease Approved 
 Certolizumab mAbs RA, Crohn disease Approved 
 Golimumab mAbs RA, PA, AS Approved 
 TNF-α kinoid Vaccine* RA, Crohn disease Phase 2 
 ESBA105 SC antibody Anterior uveitis Phase 2 
 ART621 mAbs RA, psoriasis Phase 2 
 ATN-103 Nanobody RA Phase 1 
LT-β Baminercept α LT-βR-Ig RA Phase 2 
OX40L huMAb OX40L mAbs Asthma Phase 2 
CD40L Dacetuzumab mAbs Multiple myeloma Phase 1 
   Lymphoma, large B-cell, diffuse lymphoma, NHL Phase 2 
 HCD122 mAbs Multiple myeloma Phase 1 
   Follicular lymphoma Phase 2 
FasL APO010 Fusion protein Solid tumors Phase 1 
CD27L MDX-1411 mAbs Kidney cancer Phase 1 
 MDX-1203 ADC Renal cell carcinoma, NHL Phase 1 
CD30L XmAb2513 mAbs Hodgkin lymphoma, anaplastic large-cell lymphoma Phase 1 
 SGN-35 ADC Hodgkin lymphomas Phase 3 
   Lymphoma, large-cell, anaplastic lymphoma, NHL Phase 2 
 MDX-1401 mAbs Hodgkin lymphomas Phase 1 
4–1BBL BMS-663513 mAbs Advanced cancer Phase 1 
   Melanoma Phase 2 
TRAIL Mapatumumab mAbs Colorectal, NHL, non−small-cell lung cancer Phase 2 
 Lexatumumab mAbs Kidney cancer, lymphoma, neuroblastoma, sarcoma Phase 1 
 CS-1008 mAbs Malignancies, lymphoma Phase 1 
   Colorectal neoplasms, pancreatic, lung cancer Phase 2 
RANKL Denosumab mAbs Low bone mass, low bone mineral density, osteopenia, osteoporosis Phase 3 
TWEAK BIIB023 mAbs RA Phase 1 
APRIL Atacicept Fusion protein RA Phase 2 
   SLE Phase 3 
BAFF Atacicept Fusion protein RA Phase 2 
 Benlysta mAbs Arthritis, rheumatoid, SLE Phase 2 
 LY2127399 mAbs RA Phase 2 
LIGHT Baminercept α LT-βR-Ig RA Phase 2 

RD indicates receptor derivative; SC, single-chain; PA, psoriatic arthritis; ALS, amyotrophic lateral sclerosis; AS, ankylosing spondylitis; ADC, antibody-drug conjugate; and NHL, non-Hodgkin lymphoma.

*

Vaccine composed of a keyhole limpet hemocyanin-hTNFα heterocomplex immunogen.

Recombinant fusion protein.

Ongoing study.

Several other products are currently in clinical trials. Atacicept is a human recombinant fusion protein designed to inhibit B cells, thereby suppressing autoimmune disease. This fusion protein composes the binding portion of a receptor for both BLyS and APRIL. Atacicept blocks activation of B cells by TACI, a transmembrane receptor protein found predominantly on the surface of B cells, and thus production of autoantibodies. The efficacy of atacicept in animal models of autoimmune disease and the biologic activity of atacicept in patients with SLE and RA has been demonstrated.71  Two recent phase 2 clinical trials evaluated the safety, efficacy, and biologic activity of atacicept in patients with active RA and an inadequate response to methotrexate. Despite the biologic effects of atacicept, the proportion of patients meeting the primary efficacy endpoint did not differ significantly in the atacicept groups and the placebo group.72,73  Another phase 2 clinical trial was designed to assess the safety and tolerability of atacicept and its effects on central nervous system inflammation in relapsing multiple sclerosis (IMP28063, www.clinicaltrials.gov, #NCT00642902). Surprisingly, an increase in inflammatory disease activity was reported that led to a suspension of all atacicept trials in multiple sclerosis.

One of the major problems with most of these TNF blockers is that they produce numerous side effects. For example, infliximab (Remicade), etanercept (Enbrel), and adalimumab (Humira) are immunosuppressants and carry black-box warning labels regarding the increased risk of serious infections (such as tuberculosis). These drugs also carry a warning label regarding an increased risk of central nervous system demyelinating disorders. Adverse reactions in patients receiving infliximab have been studied and reported. Risks include serious and sometimes fatal blood disorders, serious infections, lymphoma, and solid tumors. There are reports of serious liver injury, reactivation of hepatitis B, hepatosplenic T-cell lymphoma, and drug-induced lupus. Cases of leukopenia, neutropenia, thrombocytopenia, and pancytopenia (some fatal) have also been reported with infliximab. Besides safety issues, most of the TNF blockers are highly expensive. For instance, infliximab can cost as much as $22 000 a year per patient. Thus, alternatives that are safer and more affordable, yet effective, are needed.

Attempts have been made over the past several years for the identification of agents that are safe, efficacious, and inexpensive. Agents derived from natural sources have gained considerable attention because of their ability to suppress TNF expression and TNF signaling. Curcumin (diferuloylmethane) is one such agent that was isolated more than a century ago from the rhizomes of the golden spice turmeric (Curcuma longa). While searching for a TNF blocker, our group was the first to demonstrate that curcumin can block the activity of TNF-α,74  and another group showed that it can also suppress TNF production.75  Since then, we and others have shown its ability to inhibit the growth of a wide variety of tumor cells through suppression of TNF signaling. Curcumin has also been shown to inactivate TNF-α by direct binding.76  In one study, curcumin docked at the receptor-binding sites of TNF-α and exhibited direct interaction by both noncovalent and covalent interactions.77  Curcumin is currently in clinical trials for all those diseases for which TNF blockers have been approved. In addition, several other natural products are also known to suppress both the production and action of various members of the TNF superfamily.78 

From the studies described here, it is clear that TNF-α and its family members constitute a very active area of research and thus has opened up numerous possibilities for treatment of variety of human diseases, including osteoporosis, psoriasis, arthritis, and Crohn disease. The number of publications on this superfamily, their involvement in a wide variety of human diseases, their potential to treat human disease, and the interest in and sales of products based on this family all are indicators of the future potential of these agents. However, this research is just beginning and will probably continue for decades to come. It is becoming increasingly evident that, although under controlled circumstances acute inflammation has therapeutic potential, when out of control, chronic inflammation can mediate most chronic diseases. Members of the TNF superfamily are major mediators of chronic inflammation and thus need to be regulated. More understanding is required of the pathways activated by these ligands and how to interrupt these pathways selectively for the benefit of mankind.

In conclusion, the golden journey of TNF started almost 25 years ago with the discovery of the 2 proteins, discovery of their amino acid sequence, cloning of their genes, and identification of their receptors. A great deal has been learned about cell signaling by these proteins, the cellular responses, their physiologic and pathologic roles, up-regulation and down-regulation, and genetic alterations. Most of the information in hand is about TNF-α, however; and very little is known about TNF-β, which binds to the same receptor as TNF-α. For instance, is TNF-β as pro-inflammatory as TNF-α, and if not, why not? What is the true physiologic role of TRAIL? Is this cytokine designed to kill tumor cells or is its true role yet to be determined? What is the role of VEGI? How does it signal? What is the ligand for DR6 or DR3, and how do they signal? Almost 50 different proteins constitute ligands of the TNF superfamily, ligand receptors, and receptor-associated proteins. Much is yet to be learned about them, and we expect this information to be forthcoming.

The authors thank Kathryn Hale for carefully proofreading the manuscript and providing valuable comments.

This work was supported by the National Institutes of Health (Cancer Center Core grant CA-16672 and Program Project grant CA-124787-01A2) and the Center for Targeted Therapy at The University of Texas M. D. Anderson Cancer Center. B.B.A. is the Ransom Horne Jr Professor of Cancer Research.

Personal account of corresponding author: In May 1980, Dr Aggarwal left the Hormone Research Laboratory of the University of California, San Francisco, to join a start-up company in South San Francisco called Genentech. Soon after joining Genentech, he was approached by the founder and president of the company, the young entrepreneur Bob Swanson, who was obsessed with gene cloning. Bob walked into Dr Aggarwal's office and asked him to find a “cure for cancer.” When Dr Aggarwal asked how he was supposed to do that, the immediate answer of Bob was that lymphokines are a cure for cancer and if he could clone this “sucker” he'll have a cure. After extensive reading, attending a conference in Dallas, and discussions with Dr Gale Granger, a pioneer in the lymphotoxin field, Dr Aggarwal learned that lymphokines are nothing but superatants from activated lymphocytes, and were being used at the time for the treatment of cancer. With this background, Dr Aggarwal got started in an area he had known nothing about.

National Institutes of Health

Contribution: B.B.A. and J.H.K. reviewed the literature; J.H.K. wrote portions of the tables and figures; B.B.A. and S.C.G. wrote portions of the manuscript; and B.B.A. had overall editorial responsibility for the manuscript, tables, and figures.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Bharat B. Aggarwal, Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Rd, Unit 1950, Houston, TX 77054; e-mail: aggarwal@mdanderson.org.

1
Bhardwaj
 
A
Aggarwal
 
BB
Receptor-mediated choreography of life and death.
J Clin Immunol
2003
, vol. 
23
 
5
(pg. 
317
-
332
)
2
Gaur
 
U
Aggarwal
 
BB
Regulation of proliferation, survival and apoptosis by members of the TNF superfamily.
Biochem Pharmacol
2003
, vol. 
66
 
8
(pg. 
1403
-
1408
)
3
Aggarwal
 
BB
Signalling pathways of the TNF superfamily: a double-edged sword.
Nat Rev Immunol
2003
, vol. 
3
 
9
(pg. 
745
-
756
)
4
O'Malley
 
Action of bacterial polysaccharide on tumors: II. Damage of Sarcoma 37 by serum of mice treated with Serratia marcescens polysaccharide, and induced tolerance.
J Natl Cancer Inst
1962
, vol. 
29
 (pg. 
1169
-
1175
)
5
Carswell
 
EA
Old
 
LJ
Kassel
 
RL
Green
 
S
Fiore
 
N
Williamson
 
B
An endotoxin-induced serum factor that causes necrosis of tumors.
Proc Natl Acad Sci U S A
1975
, vol. 
72
 
9
(pg. 
3666
-
3670
)
6
Aggarwal
 
BB
Moffat
 
B
Harkins
 
RN
Human lymphotoxin: production by a lymphoblastoid cell line, purification, and initial characterization.
J Biol Chem
1984
, vol. 
259
 
1
(pg. 
686
-
691
)
7
Aggarwal
 
BB
Henzel
 
WJ
Moffat
 
B
Kohr
 
WJ
Harkins
 
RN
Primary structure of human lymphotoxin derived from 1788 lymphoblastoid cell line.
J Biol Chem
1985
, vol. 
260
 
4
(pg. 
2334
-
2344
)
8
Aggarwal
 
BB
Kohr
 
WJ
Hass
 
PE
, et al. 
Human tumor necrosis factor: production, purification, and characterization.
J Biol Chem
1985
, vol. 
260
 
4
(pg. 
2345
-
2354
)
9
Bringman
 
TS
Aggarwal
 
BB
Monoclonal antibodies to human tumor necrosis factors alpha and beta: application for affinity purification, immunoassays, and as structural probes.
Hybridoma
1987
, vol. 
6
 
5
(pg. 
489
-
507
)
10
Kelker
 
HC
Oppenheim
 
JD
Stone-Wolff
 
D
, et al. 
Characterization of human tumor necrosis factor produced by peripheral blood monocytes and its separation from lymphotoxin.
Int J Cancer
1985
, vol. 
36
 
1
(pg. 
69
-
73
)
11
Pennica
 
D
Nedwin
 
GE
Hayflick
 
JS
, et al. 
Human tumour necrosis factor: precursor structure, expression and homology to lymphotoxin.
Nature
1984
, vol. 
312
 
5996
(pg. 
724
-
729
)
12
Gray
 
PW
Aggarwal
 
BB
Benton
 
CV
, et al. 
Cloning and expression of cDNA for human lymphotoxin, a lymphokine with tumour necrosis activity.
Nature
1984
, vol. 
312
 
5996
(pg. 
721
-
724
)
13
Aggarwal
 
BB
Eessalu
 
TE
Hass
 
PE
Characterization of receptors for human tumour necrosis factor and their regulation by gamma-interferon.
Nature
1985
, vol. 
318
 
6047
(pg. 
665
-
667
)
14
Beutler
 
B
Greenwald
 
D
Hulmes
 
JD
, et al. 
Identity of tumour necrosis factor and the macrophage-secreted factor cachectin.
Nature
1985
, vol. 
316
 
6028
(pg. 
552
-
554
)
15
Cerami
 
A
The value of failure: the discovery of TNF and its natural inhibitor erythropoietin.
J Intern Med
2011
, vol. 
269
 
1
(pg. 
8
-
15
)
16
Schneider
 
P
Olson
 
D
Tardivel
 
A
, et al. 
Identification of a new murine tumor necrosis factor receptor locus that contains two novel murine receptors for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL).
J Biol Chem
2003
, vol. 
278
 
7
(pg. 
5444
-
5454
)
17
Moore
 
RJ
Owens
 
DM
Stamp
 
G
, et al. 
Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis.
Nat Med
1999
, vol. 
5
 
7
(pg. 
828
-
831
)
18
Balkwill
 
F
TNF-alpha in promotion and progression of cancer.
Cancer Metastasis Rev
2006
, vol. 
25
 
3
(pg. 
409
-
416
)
19
Deroose
 
JP
Eggermont
 
AM
van Geel
 
AN
, et al. 
Long-term results of tumor necrosis factor α- and melphalan-based isolated limb perfusion in locally advanced extremity soft tissue sarcomas.
J Clin Oncol
2011
, vol. 
29
 
30
(pg. 
4036
-
4044
)
20
Deroose
 
JP
Eggermont
 
AM
van Geel
 
AN
de Wilt
 
JH
Burger
 
JW
Verhoef
 
C
20 years experience of TNF-based isolated limb perfusion for in-transit melanoma metastases: TNF dose matters [published online ahead of print August 31, 2011].
Ann Surg Oncol
 
21
Grunhagen
 
DJ
de Wilt
 
JH
van Geel
 
AN
Verhoef
 
C
Eggermont
 
AM
Isolated limb perfusion with TNF-alpha and melphalan in locally advanced soft tissue sarcomas of the extremities.
Recent Results Cancer Res
2009
, vol. 
179
 (pg. 
257
-
270
)
22
Aggarwal
 
BB
Bhardwaj
 
U
Takada
 
Y
Regulation of TRAIL-induced apoptosis by ectopic expression of antiapoptotic factors.
Vitam Horm
2004
, vol. 
67
 (pg. 
453
-
483
)
23
Nikolaev
 
A
McLaughlin
 
T
O'Leary
 
DD
Tessier-Lavigne
 
M
APP binds DR6 to trigger axon pruning and neuron death via distinct caspases.
Nature
2009
, vol. 
457
 
7232
(pg. 
981
-
989
)
24
Watts
 
AD
Hunt
 
NH
Wanigasekara
 
Y
, et al. 
A casein kinase I motif present in the cytoplasmic domain of members of the tumour necrosis factor ligand family is implicated in “reverse signalling.”
EMBO J
1999
, vol. 
18
 
8
(pg. 
2119
-
2126
)
25
Harashima
 
S
Horiuchi
 
T
Hatta
 
N
, et al. 
Outside-to-inside signal through the membrane TNF-alpha induces E-selectin (CD62E) expression on activated human CD4+ T cells.
J Immunol
2001
, vol. 
166
 
1
(pg. 
130
-
136
)
26
Wiley
 
SR
Goodwin
 
RG
Smith
 
CA
Reverse signaling via CD30 ligand.
J Immunol
1996
, vol. 
157
 
8
(pg. 
3635
-
3639
)
27
Hsu
 
H
Xiong
 
J
Goeddel
 
DV
The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation.
Cell
1995
, vol. 
81
 
4
(pg. 
495
-
504
)
28
Hsu
 
H
Shu
 
HB
Pan
 
MG
Goeddel
 
DV
TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways.
Cell
1996
, vol. 
84
 
2
(pg. 
299
-
308
)
29
Morgan
 
MJ
Liu
 
ZG
Reactive oxygen species in TNFalpha-induced signaling and cell death.
Mol Cells
2010
, vol. 
30
 
1
(pg. 
1
-
12
)
30
Aggarwal
 
BB
Nuclear factor-kappaB: the enemy within.
Cancer Cell
2004
, vol. 
6
 
3
(pg. 
203
-
208
)
31
Devin
 
A
Cook
 
A
Lin
 
Y
Rodriguez
 
Y
Kelliher
 
M
Liu
 
Z
The distinct roles of TRAF2 and RIP in IKK activation by TNF-R1: TRAF2 recruits IKK to TNF-R1 while RIP mediates IKK activation.
Immunity
2000
, vol. 
12
 
4
(pg. 
419
-
429
)
32
Natoli
 
G
Costanzo
 
A
Moretti
 
F
Fulco
 
M
Balsano
 
C
Levrero
 
M
Tumor necrosis factor (TNF) receptor 1 signaling downstream of TNF receptor-associated factor 2: nuclear factor kappaB (NFkappaB)-inducing kinase requirement for activation of activating protein 1 and NFkappaB but not of c-Jun N-terminal kinase/stress-activated protein kinase.
J Biol Chem
1997
, vol. 
272
 
42
(pg. 
26079
-
26082
)
33
Ermolaeva
 
MA
Michallet
 
MC
Papadopoulou
 
N
, et al. 
Function of TRADD in tumor necrosis factor receptor 1 signaling and in TRIF-dependent inflammatory responses.
Nat Immunol
2008
, vol. 
9
 
9
(pg. 
1037
-
1046
)
34
Desch
 
CE
Dobrina
 
A
Aggarwal
 
BB
Harlan
 
JM
Tumor necrosis factor-alpha exhibits greater proinflammatory activity than lymphotoxin in vitro.
Blood
1990
, vol. 
75
 
10
(pg. 
2030
-
2034
)
35
Sugarman
 
BJ
Aggarwal
 
BB
Hass
 
PE
Figari
 
IS
Palladino
 
MA
Shepard
 
HM
Recombinant human tumor necrosis factor-alpha: effects on proliferation of normal and transformed cells in vitro.
Science
1985
, vol. 
230
 
4728
(pg. 
943
-
945
)
36
Balkwill
 
F
Tumour necrosis factor and cancer.
Nat Rev Cancer
2009
, vol. 
9
 
5
(pg. 
361
-
371
)
37
Aggarwal
 
BB
Shishodia
 
S
Sandur
 
SK
Pandey
 
MK
Sethi
 
G
Inflammation and cancer: how hot is the link?
Biochem Pharmacol
2006
, vol. 
72
 
11
(pg. 
1605
-
1621
)
38
Wiley
 
SR
Schooley
 
K
Smolak
 
PJ
, et al. 
Identification and characterization of a new member of the TNF family that induces apoptosis.
Immunity
1995
, vol. 
3
 
6
(pg. 
673
-
682
)
39
Dowlati
 
Y
Herrmann
 
N
Swardfager
 
W
, et al. 
A meta-analysis of cytokines in major depression.
Biol Psychiatry
2010
, vol. 
67
 
5
(pg. 
446
-
457
)
40
Brietzke
 
E
Kapczinski
 
F
TNF-alpha as a molecular target in bipolar disorder.
Prog Neuropsychopharmacol Biol Psychiatry
2008
, vol. 
32
 
6
(pg. 
1355
-
1361
)
41
Murashima
 
YL
Suzuki
 
J
Yoshii
 
M
Role of cytokines during epileptogenesis and in the transition from the interictal to the ictal state in the epileptic mutant EL mouse.
Gene Regul Syst Bio
2008
, vol. 
2
 (pg. 
267
-
274
)
42
Swardfager
 
W
Lanctot
 
K
Rothenburg
 
L
Wong
 
A
Cappell
 
J
Herrmann
 
N
A meta-analysis of cytokines in Alzheimer's disease.
Biol Psychiatry
2010
, vol. 
68
 
10
(pg. 
930
-
941
)
43
Nagatsu
 
T
Sawada
 
M
Inflammatory process in Parkinson's disease: role for cytokines.
Curr Pharm Des
2005
, vol. 
11
 
8
(pg. 
999
-
1016
)
44
Sayed
 
BA
Christy
 
AL
Walker
 
ME
Brown
 
MA
Meningeal mast cells affect early T cell central nervous system infiltration and blood-brain barrier integrity through TNF: a role for neutrophil recruitment?
J Immunol
2010
, vol. 
184
 
12
(pg. 
6891
-
6900
)
45
Choi
 
C
Gillespie
 
GY
Van Wagoner
 
NJ
Benveniste
 
EN
Fas engagement increases expression of interleukin-6 in human glioma cells.
J Neuro-oncol
2002
, vol. 
56
 
1
(pg. 
13
-
19
)
46
Kapadia
 
S
Lee
 
J
Torre-Amione
 
G
Birdsall
 
HH
Ma
 
TS
Mann
 
DL
Tumor necrosis factor-alpha gene and protein expression in adult feline myocardium after endotoxin administration.
J Clin Invest
1995
, vol. 
96
 
2
(pg. 
1042
-
1052
)
47
Levine
 
B
Kalman
 
J
Mayer
 
L
Fillit
 
HM
Packer
 
M
Elevated circulating levels of tumor necrosis factor in severe chronic heart failure.
N Engl J Med
1990
, vol. 
323
 
4
(pg. 
236
-
241
)
48
Feldman
 
AM
Combes
 
A
Wagner
 
D
, et al. 
The role of tumor necrosis factor in the pathophysiology of heart failure.
J Am Coll Cardiol
2000
, vol. 
35
 
3
(pg. 
537
-
544
)
49
Diwan
 
A
Tran
 
T
Misra
 
A
Mann
 
DL
Inflammatory mediators and the failing heart: a translational approach.
Curr Mol Med
2003
, vol. 
3
 
2
(pg. 
161
-
182
)
50
McKellar
 
GE
McCarey
 
DW
Sattar
 
N
McInnes
 
IB
Role for TNF in atherosclerosis? Lessons from autoimmune disease.
Nat Rev Cardiol
2009
, vol. 
6
 
6
(pg. 
410
-
417
)
51
Avina-Zubieta
 
JA
Choi
 
HK
Sadatsafavi
 
M
Etminan
 
M
Esdaile
 
JM
Lacaille
 
D
Risk of cardiovascular mortality in patients with rheumatoid arthritis: a meta-analysis of observational studies.
Arthritis Rheum
2008
, vol. 
59
 
12
(pg. 
1690
-
1697
)
52
Matera
 
MG
Calzetta
 
L
Cazzola
 
M
TNF-alpha inhibitors in asthma and COPD: we must not throw the baby out with the bath water.
Pulm Pharmacol Ther
2010
, vol. 
23
 
2
(pg. 
121
-
128
)
53
Kips
 
JC
Tavernier
 
JH
Joos
 
GF
Peleman
 
RA
Pauwels
 
RA
The potential role of tumour necrosis factor alpha in asthma.
Clin Exp Allergy
1993
, vol. 
23
 
4
(pg. 
247
-
250
)
54
Lundblad
 
LK
Thompson-Figueroa
 
J
Leclair
 
T
, et al. 
Tumor necrosis factor-alpha overexpression in lung disease: a single cause behind a complex phenotype.
Am J Respir Crit Care Med
2005
, vol. 
171
 
12
(pg. 
1363
-
1370
)
55
Collison
 
A
Foster
 
PS
Mattes
 
J
Emerging role of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) as a key regulator of inflammatory responses.
Clin Exp Pharmacol Physiol
2009
, vol. 
36
 
11
(pg. 
1049
-
1053
)
56
Schreiber
 
TH
Wolf
 
D
Tsai
 
MS
, et al. 
Therapeutic Treg expansion in mice by TNFRSF25 prevents allergic lung inflammation.
J Clin Invest
2010
, vol. 
120
 
10
(pg. 
3629
-
3640
)
57
Hotamisligil
 
GS
Shargill
 
NS
Spiegelman
 
BM
Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance.
Science
1993
, vol. 
259
 
5091
(pg. 
87
-
91
)
58
Tzanavari
 
T
Giannogonas
 
P
Karalis
 
KP
TNF-alpha and obesity.
Curr Dir Autoimmun
2010
, vol. 
11
 (pg. 
145
-
156
)
59
Dandona
 
P
Aljada
 
A
Bandyopadhyay
 
A
Inflammation: the link between insulin resistance, obesity and diabetes.
Trends Immunol
2004
, vol. 
25
 
1
(pg. 
4
-
7
)
60
Vinay
 
DS
Kwon
 
BS
The tumour necrosis factor/TNF receptor superfamily: therapeutic targets in autoimmune diseases.
Clin Exp Immunol
2011
, vol. 
164
 (pg. 
145
-
157
)
61
Banks
 
TA
Rouse
 
BT
Kerley
 
MK
, et al. 
Lymphotoxin-alpha-deficient mice: effects on secondary lymphoid organ development and humoral immune responsiveness.
J Immunol
1995
, vol. 
155
 
4
(pg. 
1685
-
1693
)
62
Pasparakis
 
M
Alexopoulou
 
L
Episkopou
 
V
Kollias
 
G
Immune and inflammatory responses in TNF alpha-deficient mice: a critical requirement for TNF alpha in the formation of primary B cell follicles, follicular dendritic cell networks and germinal centers, and in the maturation of the humoral immune response.
J Exp Med
1996
, vol. 
184
 
4
(pg. 
1397
-
1411
)
63
Marino
 
MW
Dunn
 
A
Grail
 
D
, et al. 
Characterization of tumor necrosis factor-deficient mice.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
15
(pg. 
8093
-
8098
)
64
Hubert
 
KE
Davies
 
MH
Stempel
 
AJ
Griffith
 
TS
Powers
 
MR
TRAIL-deficient mice exhibit delayed regression of retinal neovascularization.
Am J Pathol
2009
, vol. 
175
 
6
(pg. 
2697
-
2708
)
65
Lamhamedi-Cherradi
 
SE
Zheng
 
SJ
Maguschak
 
KA
Peschon
 
J
Chen
 
YH
Defective thymocyte apoptosis and accelerated autoimmune diseases in TRAIL−/− mice.
Nat Immunol
2003
, vol. 
4
 
3
(pg. 
255
-
260
)
66
Finnberg
 
N
Gruber
 
JJ
Fei
 
P
, et al. 
DR5 knockout mice are compromised in radiation-induced apoptosis.
Mol Cell Biol
2005
, vol. 
25
 
5
(pg. 
2000
-
2013
)
67
Ida
 
H
Kawasaki
 
E
Miyashita
 
T
, et al. 
A novel mutation (T61I) in the gene encoding tumour necrosis factor receptor superfamily 1A (TNFRSF1A) in a Japanese patient with tumour necrosis factor receptor-associated periodic syndrome (TRAPS) associated with systemic lupus erythematosus.
Rheumatology (Oxford)
2004
, vol. 
43
 
10
(pg. 
1292
-
1299
)
68
Um
 
JY
An
 
NH
Kim
 
HM
TNF-alpha and TNF-beta gene polymorphisms in cerebral infarction.
J Mol Neurosci
2003
, vol. 
21
 
2
(pg. 
167
-
171
)
69
Boyce
 
EG
Halilovic
 
J
Stan-Ugbene
 
O
Golimumab: review of the efficacy and tolerability of a recently approved tumor necrosis factor-alpha inhibitor.
Clin Ther
2010
, vol. 
32
 
10
(pg. 
1681
-
1703
)
70
Schreiber
 
S
Rutgeerts
 
P
Fedorak
 
RN
, et al. 
A randomized, placebo-controlled trial of certolizumab pegol (CDP870) for treatment of Crohn's disease.
Gastroenterology
2005
, vol. 
129
 
3
(pg. 
807
-
818
)
71
Hartung
 
HP
Kieseier
 
BC
Atacicept: targeting B cells in multiple sclerosis.
Ther Adv Neurol Disord
2010
, vol. 
3
 
4
(pg. 
205
-
216
)
72
Genovese
 
MC
Kinnman
 
N
de La Bourdonnaye
 
G
Pena Rossi
 
C
Tak
 
PP
Atacicept in patients with rheumatoid arthritis and an inadequate response to tumor necrosis factor antagonist therapy: results of a phase II, randomized, placebo-controlled, dose-finding trial.
Arthritis Rheum
2011
, vol. 
63
 
7
(pg. 
1793
-
1803
)
73
van Vollenhoven
 
RF
Kinnman
 
N
Vincent
 
E
Wax
 
S
Bathon
 
J
Atacicept in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase II, randomized, placebo-controlled trial.
Arthritis Rheum
2011
, vol. 
63
 
7
(pg. 
1782
-
1792
)
74
Singh
 
S
Aggarwal
 
BB
Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane) [corrected].
J Biol Chem
1995
, vol. 
270
 
42
(pg. 
24995
-
25000
)
75
Chan
 
MM
Inhibition of tumor necrosis factor by curcumin, a phytochemical.
Biochem Pharmacol
1995
, vol. 
49
 
11
(pg. 
1551
-
1556
)
76
Gupta
 
SC
Prasad
 
S
Kim
 
JH
, et al. 
Multitargeting by curcumin as revealed by molecular interaction studies
Nat Prod Rep
2011
, vol. 
28
 
12
(pg. 
1937
-
1955
)
77
Wua
 
ST
Suna
 
JC
Leeb
 
KJ
Sunc
 
YM
Docking prediction for tumor necrosis factor-α and five herbal inhibitors.
Intl J Eng Sci Technol
2010
, vol. 
2
 (pg. 
4263
-
4277
)
78
Sethi
 
G
Sung
 
B
Aggarwal
 
BB
TNF: a master switch for inflammation to cancer.
Front Biosci
2008
, vol. 
13
 (pg. 
5094
-
5107
)
79
Song
 
HY
Regnier
 
CH
Kirschning
 
CJ
Goeddel
 
DV
Rothe
 
M
Tumor necrosis factor (TNF)-mediated kinase cascades: bifurcation of nuclear factor-kappaB and c-jun N-terminal kinase (JNK/SAPK) pathways at TNF receptor-associated factor 2.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
18
(pg. 
9792
-
9796
)
80
Yuasa
 
T
Ohno
 
S
Kehrl
 
JH
Kyriakis
 
JM
Tumor necrosis factor signaling to stress-activated protein kinase (SAPK)/Jun NH2-terminal kinase (JNK) and p38: germinal center kinase couples TRAF2 to mitogen-activated protein kinase/ERK kinase kinase 1 and SAPK while receptor interacting protein associates with a mitogen-activated protein kinase kinase kinase upstream of MKK6 and p38.
J Biol Chem
1998
, vol. 
273
 
35
(pg. 
22681
-
22692
)
81
Alimzhanov
 
MB
Kuprash
 
DV
Kosco-Vilbois
 
MH
, et al. 
Abnormal development of secondary lymphoid tissues in lymphotoxin beta-deficient mice.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
17
(pg. 
9302
-
9307
)
82
Locksley
 
RM
Killeen
 
N
Lenardo
 
MJ
The TNF and TNF receptor superfamilies: integrating mammalian biology.
Cell
2001
, vol. 
104
 
4
(pg. 
487
-
501
)
83
Kim
 
N
Odgren
 
PR
Kim
 
DK
Marks
 
SC
Choi
 
Y
Diverse roles of the tumor necrosis factor family member TRANCE in skeletal physiology revealed by TRANCE deficiency and partial rescue by a lymphocyte-expressed TRANCE transgene.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 
20
(pg. 
10905
-
10910
)
84
Varfolomeev
 
E
Kischkel
 
F
Martin
 
F
, et al. 
APRIL-deficient mice have normal immune system development.
Mol Cell Biol
2004
, vol. 
24
 
3
(pg. 
997
-
1006
)
85
Castigli
 
E
Scott
 
S
Dedeoglu
 
F
, et al. 
Impaired IgA class switching in APRIL-deficient mice.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
11
(pg. 
3903
-
3908
)
86
Gross
 
JA
Dillon
 
SR
Mudri
 
S
, et al. 
TACI-Ig neutralizes molecules critical for B cell development and autoimmune disease. impaired B cell maturation in mice lacking BLyS.
Immunity
2001
, vol. 
15
 
2
(pg. 
289
-
302
)
87
Schiemann
 
B
Gommerman
 
JL
Vora
 
K
, et al. 
An essential role for BAFF in the normal development of B cells through a BCMA-independent pathway.
Science
2001
, vol. 
293
 
5537
(pg. 
2111
-
2114
)
88
Monreal
 
AW
Zonana
 
J
Ferguson
 
B
Identification of a new splice form of the EDA1 gene permits detection of nearly all X-linked hypohidrotic ectodermal dysplasia mutations.
Am J Hum Genet
1998
, vol. 
63
 
2
(pg. 
380
-
389
)
89
Schneider
 
P
Street
 
SL
Gaide
 
O
, et al. 
Mutations leading to X-linked hypohidrotic ectodermal dysplasia affect three major functional domains in the tumor necrosis factor family member ectodysplasin-A.
J Biol Chem
2001
, vol. 
276
 
22
(pg. 
18819
-
18827
)
90
Newton
 
K
French
 
DM
Yan
 
M
Frantz
 
GD
Dixit
 
VM
Myodegeneration in EDA-A2 transgenic mice is prevented by XEDAR deficiency.
Mol Cell Biol
2004
, vol. 
24
 
4
(pg. 
1608
-
1613
)
91
Rothe
 
J
Lesslauer
 
W
Lotscher
 
H
, et al. 
Mice lacking the tumour necrosis factor receptor 1 are resistant to TNF-mediated toxicity but highly susceptible to infection by Listeria monocytogenes.
Nature
1993
, vol. 
364
 
6440
(pg. 
798
-
802
)
92
Knight
 
B
Yeoh
 
GC
Husk
 
KL
, et al. 
Impaired preneoplastic changes and liver tumor formation in tumor necrosis factor receptor type 1 knockout mice.
J Exp Med
2000
, vol. 
192
 
12
(pg. 
1809
-
1818
)
93
Rieux-Laucat
 
F
Le Deist
 
F
Hivroz
 
C
, et al. 
Mutations in Fas associated with human lymphoproliferative syndrome and autoimmunity.
Science
1995
, vol. 
268
 
5215
(pg. 
1347
-
1349
)
94
Jacobson
 
BA
Panka
 
DJ
Nguyen
 
KA
Erikson
 
J
Abbas
 
AK
Marshak-Rothstein
 
A
Anatomy of autoantibody production: dominant localization of antibody-producing cells to T cell zones in Fas-deficient mice.
Immunity
1995
, vol. 
3
 
4
(pg. 
509
-
519
)
95
Singer
 
GG
Abbas
 
AK
The fas antigen is involved in peripheral but not thymic deletion of T lymphocytes in T cell receptor transgenic mice.
Immunity
1994
, vol. 
1
 
5
(pg. 
365
-
371
)
96
Gaspal
 
FM
Kim
 
MY
McConnell
 
FM
Raykundalia
 
C
Bekiaris
 
V
Lane
 
PJ
Mice deficient in OX40 and CD30 signals lack memory antibody responses because of deficient CD4 T cell memory.
J Immunol
2005
, vol. 
174
 
7
(pg. 
3891
-
3896
)
97
Kwon
 
BS
Hurtado
 
JC
Lee
 
ZH
, et al. 
Immune responses in 4-1BB (CD137)-deficient mice.
J Immunol
2002
, vol. 
168
 
11
(pg. 
5483
-
5490
)
98
Vinay
 
DS
Choi
 
BK
Bae
 
JS
Kim
 
WY
Gebhardt
 
BM
Kwon
 
BS
CD137-deficient mice have reduced NK/NKT cell numbers and function, are resistant to lipopolysaccharide-induced shock syndromes, and have lower IL-4 responses.
J Immunol
2004
, vol. 
173
 
6
(pg. 
4218
-
4229
)
99
Lee
 
SW
Park
 
Y
So
 
T
, et al. 
Identification of regulatory functions for 4-1BB and 4-1BBL in myelopoiesis and the development of dendritic cells.
Nat Immunol
2008
, vol. 
9
 
8
(pg. 
917
-
926
)
100
Jeon
 
HJ
Choi
 
JH
Jung
 
IH
, et al. 
CD137 (4-1BB) deficiency reduces atherosclerosis in hyperlipidemic mice.
Circulation
2010
, vol. 
121
 
9
(pg. 
1124
-
1133
)
101
Girgenrath
 
M
Weng
 
S
Kostek
 
CA
, et al. 
TWEAK, via its receptor Fn14, is a novel regulator of mesenchymal progenitor cells and skeletal muscle regeneration.
EMBO J
2006
, vol. 
25
 
24
(pg. 
5826
-
5839
)
102
Scholzke
 
MN
Rottinger
 
A
Murikinati
 
S
Gehrig
 
N
Leib
 
C
Schwaninger
 
M
TWEAK regulates proliferation and differentiation of adult neural progenitor cells.
Mol Cell Neurosci
2011
, vol. 
46
 
1
(pg. 
325
-
332
)
103
Tirnitz-Parker
 
JE
Viebahn
 
CS
Jakubowski
 
A
, et al. 
Tumor necrosis factor-like weak inducer of apoptosis is a mitogen for liver progenitor cells.
Hepatology
2010
, vol. 
52
 
1
(pg. 
291
-
302
)
104
Yan
 
M
Wang
 
H
Chan
 
B
, et al. 
Activation and accumulation of B cells in TACI-deficient mice.
Nat Immunol
2001
, vol. 
2
 
7
(pg. 
638
-
643
)
105
Yan
 
M
Brady
 
JR
Chan
 
B
, et al. 
Identification of a novel receptor for B lymphocyte stimulator that is mutated in a mouse strain with severe B cell deficiency.
Curr Biol
2001
, vol. 
11
 
19
(pg. 
1547
-
1552
)
106
Wang
 
EC
Thern
 
A
Denzel
 
A
Kitson
 
J
Farrow
 
SN
Owen
 
MJ
DR3 regulates negative selection during thymocyte development.
Mol Cell Biol
2001
, vol. 
21
 
10
(pg. 
3451
-
3461
)
107
Stephens
 
GL
McHugh
 
RS
Whitters
 
MJ
, et al. 
Engagement of glucocorticoid-induced TNFR family-related receptor on effector T cells by its ligand mediates resistance to suppression by CD4+CD25+ T cells.
J Immunol
2004
, vol. 
173
 
8
(pg. 
5008
-
5020
)
108
Pispa
 
J
Pummila
 
M
Barker
 
PA
Thesleff
 
I
Mikkola
 
ML
Edar and Troy signalling pathways act redundantly to regulate initiation of hair follicle development.
Hum Mol Genet
2008
, vol. 
17
 
21
(pg. 
3380
-
3391
)
109
Schmidt
 
CS
Liu
 
J
Zhang
 
T
, et al. 
Enhanced B cell expansion, survival, and humoral responses by targeting death receptor 6.
J Exp Med
2003
, vol. 
197
 
1
(pg. 
51
-
62
)
110
Zhao
 
H
Yan
 
M
Wang
 
H
Erickson
 
S
Grewal
 
IS
Dixit
 
VM
Impaired c-Jun amino terminal kinase activity and T cell differentiation in death receptor 6-deficient mice.
J Exp Med
2001
, vol. 
194
 
10
(pg. 
1441
-
1448
)
111
Liu
 
J
Na
 
S
Glasebrook
 
A
, et al. 
Enhanced CD4+ T cell proliferation and Th2 cytokine production in DR6-deficient mice.
Immunity
2001
, vol. 
15
 
1
(pg. 
23
-
34
)
112
Lee
 
KF
Li
 
E
Huber
 
LJ
, et al. 
Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system.
Cell
1992
, vol. 
69
 
5
(pg. 
737
-
749
)
113
Tsitsikov
 
EN
Laouini
 
D
Dunn
 
IF
, et al. 
TRAF1 is a negative regulator of TNF signaling. enhanced TNF signaling in TRAF1-deficient mice.
Immunity
2001
, vol. 
15
 
4
(pg. 
647
-
657
)
114
Missiou
 
A
Kostlin
 
N
Varo
 
N
, et al. 
Tumor necrosis factor receptor-associated factor 1 (TRAF1) deficiency attenuates atherosclerosis in mice by impairing monocyte recruitment to the vessel wall.
Circulation
2010
, vol. 
121
 
18
(pg. 
2033
-
2044
)
115
Nguyen
 
LT
Duncan
 
GS
Mirtsos
 
C
, et al. 
TRAF2 deficiency results in hyperactivity of certain TNFR1 signals and impairment of CD40-mediated responses.
Immunity
1999
, vol. 
11
 
3
(pg. 
379
-
389
)
116
Xu
 
Y
Cheng
 
G
Baltimore
 
D
Targeted disruption of TRAF3 leads to postnatal lethality and defective T-dependent immune responses.
Immunity
1996
, vol. 
5
 
5
(pg. 
407
-
415
)
117
Nakano
 
H
Sakon
 
S
Koseki
 
H
, et al. 
Targeted disruption of Traf5 gene causes defects in CD40- and CD27-mediated lymphocyte activation.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 
17
(pg. 
9803
-
9808
)
118
Lomaga
 
MA
Yeh
 
WC
Sarosi
 
I
, et al. 
TRAF6 deficiency results in osteopetrosis and defective interleukin-1, CD40, and LPS signaling.
Genes Dev
1999
, vol. 
13
 
8
(pg. 
1015
-
1024
)
119
Naito
 
A
Yoshida
 
H
Nishioka
 
E
, et al. 
TRAF6-deficient mice display hypohidrotic ectodermal dysplasia.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 
13
(pg. 
8766
-
8771
)
120
Goeddel
 
DV
Signal transduction by tumor necrosis factor: the Parker B. Francis Lectureship.
Chest
1999
, vol. 
116
 
1 suppl
(pg. 
69S
-
73S
)
121
Yeh
 
WC
Pompa
 
JL
McCurrach
 
ME
, et al. 
FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis.
Science
1998
, vol. 
279
 
5358
(pg. 
1954
-
1958
)
122
Kreder
 
D
Krut
 
O
Adam-Klages
 
S
, et al. 
Impaired neutral sphingomyelinase activation and cutaneous barrier repair in FAN-deficient mice.
EMBO J
1999
, vol. 
18
 
9
(pg. 
2472
-
2479
)
123
Varfolomeev
 
EE
Schuchmann
 
M
Luria
 
V
, et al. 
Targeted disruption of the mouse Caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally.
Immunity
1998
, vol. 
9
 
2
(pg. 
267
-
276
)
124
Kelliher
 
MA
Grimm
 
S
Ishida
 
Y
Kuo
 
F
Stanger
 
BZ
Leder
 
P
The death domain kinase RIP mediates the TNF-induced NF-kappaB signal.
Immunity
1998
, vol. 
8
 
3
(pg. 
297
-
303
)
125
Hu
 
Y
Baud
 
V
Delhase
 
M
, et al. 
Abnormal morphogenesis but intact IKK activation in mice lacking the IKKalpha subunit of IkappaB kinase.
Science
1999
, vol. 
284
 
5412
(pg. 
316
-
320
)
126
Li
 
ZW
Chu
 
W
Hu
 
Y
, et al. 
The IKKbeta subunit of IkappaB kinase (IKK) is essential for nuclear factor kappaB activation and prevention of apoptosis.
J Exp Med
1999
, vol. 
189
 
11
(pg. 
1839
-
1845
)
127
Yoshida
 
H
Kong
 
YY
Yoshida
 
R
, et al. 
Apaf1 is required for mitochondrial pathways of apoptosis and brain development.
Cell
1998
, vol. 
94
 
6
(pg. 
739
-
750
)
128
Hakem
 
R
Hakem
 
A
Duncan
 
GS
, et al. 
Differential requirement for caspase 9 in apoptotic pathways in vivo.
Cell
1998
, vol. 
94
 
3
(pg. 
339
-
352
)
129
Kuida
 
K
Zheng
 
TS
Na
 
S
, et al. 
Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice.
Nature
1996
, vol. 
384
 
6607
(pg. 
368
-
372
)
130
Headon
 
DJ
Emmal
 
SA
Ferguson
 
BM
, et al. 
Gene defect in ectodermal dysplasia implicates a death domain adapter in development.
Nature
2001
, vol. 
414
 
6866
(pg. 
913
-
916
)
131
Li
 
X
Act1 modulates autoimmunity through its dual functions in CD40L/BAFF and IL-17 signaling.
Cytokine
2008
, vol. 
41
 
2
(pg. 
105
-
113
)
132
Allen
 
RC
Armitage
 
RJ
Conley
 
ME
, et al. 
CD40 ligand gene defects responsible for X-linked hyper-IgM syndrome.
Science
1993
, vol. 
259
 
5097
(pg. 
990
-
993
)
133
Takahashi
 
T
Tanaka
 
M
Brannan
 
CI
, et al. 
Generalized lymphoproliferative disease in mice, caused by a point mutation in the Fas ligand.
Cell
1994
, vol. 
76
 
6
(pg. 
969
-
976
)
134
Casal
 
ML
Scheidt
 
JL
Rhodes
 
JL
Henthorn
 
PS
Werner
 
P
Mutation identification in a canine model of X-linked ectodermal dysplasia.
Mamm Genome
2005
, vol. 
16
 
7
(pg. 
524
-
531
)
135
McDermott
 
MF
Aksentijevich
 
I
Galon
 
J
, et al. 
Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes.
Cell
1999
, vol. 
97
 
1
(pg. 
133
-
144
)
136
Waschke
 
KA
Villani
 
AC
Vermeire
 
S
, et al. 
Tumor necrosis factor receptor gene polymorphisms in Crohn's disease: association with clinical phenotypes.
Am J Gastroenterol
2005
, vol. 
100
 
5
(pg. 
1126
-
1133
)
137
Magerus-Chatinet
 
A
Neven
 
B
Stolzenberg
 
MC
, et al. 
Onset of autoimmune lymphoproliferative syndrome (ALPS) in humans as a consequence of genetic defect accumulation.
J Clin Invest
2011
, vol. 
121
 
1
(pg. 
106
-
112
)
138
Hughes
 
AE
Ralston
 
SH
Marken
 
J
, et al. 
Mutations in TNFRSF11A, affecting the signal peptide of RANK, cause familial expansile osteolysis.
Nat Genet
2000
, vol. 
24
 
1
(pg. 
45
-
48
)
139
Cundy
 
T
Hegde
 
M
Naot
 
D
, et al. 
A mutation in the gene TNFRSF11B encoding osteoprotegerin causes an idiopathic hyperphosphatasia phenotype.
Hum Mol Genet
2002
, vol. 
11
 
18
(pg. 
2119
-
2127
)
140
Middleton-Hardie
 
C
Zhu
 
Q
Cundy
 
H
, et al. 
Deletion of aspartate 182 in OPG causes juvenile Paget's disease by impairing both protein secretion and binding to RANKL.
J Bone Miner Res
2006
, vol. 
21
 
3
(pg. 
438
-
445
)
141
Salzer
 
U
Chapel
 
HM
Webster
 
AD
, et al. 
Mutations in TNFRSF13B encoding TACI are associated with common variable immunodeficiency in humans.
Nat Genet
2005
, vol. 
37
 
8
(pg. 
820
-
828
)
142
Mayne
 
CG
Amanna
 
IJ
Nashold
 
FE
Hayes
 
CE
Systemic autoimmunity in BAFF-R-mutant A/WySnJ strain mice.
Eur J Immunol
2008
, vol. 
38
 
2
(pg. 
587
-
598
)
143
Shimomura
 
Y
Sato
 
N
Miyashita
 
A
Hashimoto
 
T
Ito
 
M
Kuwano
 
R
A rare case of hypohidrotic ectodermal dysplasia caused by compound heterozygous mutations in the EDAR gene.
J Invest Dermatol
2004
, vol. 
123
 
4
(pg. 
649
-
655
)
144
Perez de Diego
 
R
Sancho-Shimizu
 
V
Lorenzo
 
L
, et al. 
Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like receptor 3 response and susceptibility to herpes simplex encephalitis.
Immunity
2010
, vol. 
33
 
3
(pg. 
400
-
411
)
Sign in via your Institution