Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Signalling pathways of the TNF superfamily: a double-edged sword

Key Points

  • 19 ligands and 29 receptors that belong to the tumour-necrosis factor (TNF) superfamily have been identified.

  • At the cellular level TNF-superfamily members promote either apoptosis, proliferation, survival or differentiation.

  • The main signals transduced by the TNF-superfamily members include activation of nuclear factor-κB (NF-κB), JUN N-terminal kinase, p38 mitogen activated protein kinase and ERK1/ERK2.

  • Members of the TNF superfamily mediate haematopoiesis, immune surveillance, tumour regression and protection from infection.

  • Members of the TNF superfamily mediate inflammation, autoimmune diseases, rheumatoid arthritis, tumour metastasis, septic shock and osteoporosis.

  • TNF and its inhibitors have been approved as therapeutics.

Abstract

Two different tumour-necrosis factors (TNFs), first isolated in 1984, were found to be cytotoxic to tumour cells and to induce tumour regression in mice. Research during the past two decades has shown the existence of a superfamily of TNF proteins consisting of 19 members that signal through 29 receptors. These ligands, while regulating normal functions such as immune responses, haematopoiesis and morphogenesis, have also been implicated in tumorigenesis, transplant rejection, septic shock, viral replication, bone resorption, rheumatoid arthritis and diabetes; so indicating their role as 'double-edged swords'. These cytokines either induce cellular proliferation, survival, differentiation or apoptosis. Blockers of TNF have been approved for human use in treating TNF-linked autoimmune diseases in the United States and other countries.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: A diagrammatic representation of the ligands of the TNF superfamily and their receptors.
Figure 2: Cellular signalling pathways leading to activation of the main cellular responses by members of the TNF superfamily.
Figure 3: The structures of different TNF receptor-associated factors.
Figure 4: The balance between life and death mediated by TNF-superfamily members.
Figure 5: The main physiological and pathological effects linked to members of the TNF superfamily.
Figure 6: Present and future therapeutics.

Similar content being viewed by others

References

  1. Bruns, P. Die Heilwirkung des Erysipels auf Geschwulste. Beitr. Klin. Chir. 3, 443–446 (1868).

    Google Scholar 

  2. Coley, W. B. Contribution to the knowledge of Sarcoma. Ann. Surg. 14, 199–220 (1891).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gratia, A. & Linz, R. C. R. Seances Soc. Biol. Ses Fil. 108, 421–428 (1931).

    Google Scholar 

  4. Shear, M. J. & Turner, F. C. Chemical treatment of tumors. V. Isolation of the hemorrhage-producing fraction from Serratia marcescens (Bacillus prodigiosus) culture filtrate. J. Natl Cancer Inst. 4, 81–97 (1943).

    CAS  Google Scholar 

  5. Algire, G. H., Legallaies, F. Y. & Anderson, B. F. Vascular reactions of normal and malignant tissues in vivo. V. The role of hypotension in the action of a bacterial polysaccharide on tumors. J. Natl Cancer Inst. 12, 1279–1295 (1952).

    CAS  PubMed  Google Scholar 

  6. O'Malley, W. E., Achinstein, B. & Shear, M. J. 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. 29, 1169–1175 (1962).

    CAS  Google Scholar 

  7. Carswell, E. A. et al. An endotoxin-induced serum factor that causes necrosis of tumors. Proc. Natl Acad. Sci. USA 72, 3666–3670 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Williamson, B. D. et al. Human tumor necrosis factor produced by human B-cell lines: synergistic cytotoxic interaction with human interferon. Proc. Natl Acad. Sci. USA 80, 5397–5401 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Williams, T. W. et al. Lymphocyte in vitro cytotoxicity: lymphotoxins of several mammalian species. Nature 7, 1076–1077 (1968).

    Article  Google Scholar 

  10. Aggarwal, B. B. et al. Human lymphotoxin: production by a lymphoblastoid cell line, purification and initial characterization. J. Biol. Chem. 259, 686–691 (1984). This is the first report on the isolation and characterization of lymphotoxin (LT) or tumour-necrosis factor-β (TNF-β).

    CAS  PubMed  Google Scholar 

  11. Aggarwal, B. B. et al. Primary structure of human lymphotoxin derived from 1788 lymphoblastoid cell line. J. Biol. Chem. 260, 2334–2344 (1985).

    CAS  PubMed  Google Scholar 

  12. Aggarwal, B. B. et al. Human tumor necrosis factor: production, purification and characterization. J. Biol. Chem. 260, 2345–2354 (1985). This is the first report on the isolation and characterization of TNF-α.

    CAS  PubMed  Google Scholar 

  13. Gray, P. W. et al. Cloning and expression of cDNA for human lymphotoxin, a lymphokine with tumor necrosis activity. Nature 312, 721–724 (1984).

    Article  CAS  PubMed  Google Scholar 

  14. Pennica, D. et al. Human tumor necrosis factor: precursor structure, expression and homology to lymphotoxin. Nature 312, 724–729 (1984).

    Article  CAS  PubMed  Google Scholar 

  15. Aggarwal, B. B. et al. Characterization of receptors for human tumor necrosis factor and their regulation by γ-interferon. Nature 318, 665–667 (1985). This is the first report to describe the characterization of TNF receptors (TNFRs) and indicates that LT and TNF have a common receptor.

    Article  CAS  PubMed  Google Scholar 

  16. Beutler, B. et al. Identity of tumour necrosis factor and the macrophage-secreted factor cachectin. Nature 316, 552–554 (1985).

    Article  CAS  PubMed  Google Scholar 

  17. Takeda, K. et al. Identity of differentiation inducing factor and tumour necrosis factor. Nature 323, 338–340 (1986).

    Article  CAS  PubMed  Google Scholar 

  18. Smith, C. A. et al. The TNF receptor superfamily of cellular and viral proteins: activation, co–stimulation, and death. Cell 76, 959–962 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Locksley, R. M. et al. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104, 487–501 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Yonehara, S. et al. A cell-killing monoclonal antibody (anti-Fas) to a cell surface antigen co-downregulated with the receptor of tumor necrosis factor. J. Exp. Med. 169, 1747–1756 (1989).

    Article  CAS  PubMed  Google Scholar 

  21. Itoh, N. et al. The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis. Cell 66, 233–243 (1991).

    Article  CAS  PubMed  Google Scholar 

  22. Wiley, S. R. et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity 3, 673–682 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. Pitti, R. M. et al. Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J. Biol. Chem. 271, 12687–12690 (1996).

    Article  CAS  PubMed  Google Scholar 

  24. Anderson, D. M. et al. A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 390, 175–179 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Wong, B. R. et al. TRANCE (tumor necrosis factor (TNF)-related activation-induced cytokine), a new TNF family member predominantly expressed in T cells, is a dendritic cell-specific survival factor. J. Exp. Med. 186, 2075–2080 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lacey, D. L. et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93, 165–176 (1998).

    Article  CAS  PubMed  Google Scholar 

  27. Haridas, V. et al. VEGI, a new member of the TNF family activates nuclear factor-κB and c-Jun N-terminal kinase and modulates cell growth. Oncogene 18, 6496–6504 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Migone, T. S. et al. TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell co-stimulator. Immunity 16, 479–492 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Mukhopadhyay, A. et al. Identification and characterization of a novel cytokine, THANK, a TNF homologue that activates apoptosis, nuclear factor-κB, and c-Jun NH2-terminal kinase. J. Biol. Chem. 274, 15978–15981 (1999).

    Article  CAS  PubMed  Google Scholar 

  30. Schneider, P. et al. BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth. J. Exp. Med. 189, 1747–1756 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Nocentini, G. et al. A new member of the tumor necrosis factor/nerve growth factor receptor family inhibits T cell receptor-induced apoptosis. Proc. Natl Acad. Sci. USA 94, 6216–6221 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Black, R. A. et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-α from cells. Nature 385, 729–733 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Lum, L. et al. Evidence for a role of a tumor necrosis factor-α (TNF-α)-converting enzyme–like protease in shedding of TRANCE, a TNF family member involved in osteoclastogenesis and dendritic cell survival. J. Biol. Chem. 274, 13613–13618 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Powell, W. C. et al. The metalloproteinase matrilysin proteolytically generates active soluble Fas ligand and potentiates epithelial cell apoptosis. Curr. Biol. 9, 1441–1447 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Chen, Y. et al. Mutations within a furin consensus sequence block proteolytic release of ectodysplasin-A and cause X-linked hypohidrotic ectodermal dysplasia. Proc. Natl Acad. Sci. USA 98, 7218–7223 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Suda, T. et al. Membrane Fas ligand kills human peripheral blood T lymphocytes, and soluble Fas ligand blocks the killing. J. Exp. Med. 186, 2045–2050 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. McDermott, M. F. et al. Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell 97, 133–144 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. Simonet, W. S. et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 89, 309–319 (1997).

    Article  CAS  PubMed  Google Scholar 

  39. Itoh, N. et al. A novel protein domain required for apoptosis. Mutational analysis of human Fas antigen. J. Biol. Chem. 268, 10932–10937 (1993). This report and reference 40 were the first to describe the presence of a death domain in TNFR and CD95.

    CAS  PubMed  Google Scholar 

  40. Tartaglia, L. A. et al. A novel domain within the 55 kd TNF receptor signals cell death. Cell 74, 845–853 (1993).

    Article  CAS  PubMed  Google Scholar 

  41. Becraft, P. W. et al. CRINKLY4: a TNFR-like receptor kinase involved in maize epidermal differentiation. Science 273, 1406–1409 (1996).

    Article  CAS  PubMed  Google Scholar 

  42. Chan, F. K. et al. A domain in TNF receptors that mediates ligand-independent receptor assembly and signaling. Science 288, 2351–2354 (2000). This report refutes the previous theory proposing that the ligand induces oligomerization of the receptor. The authors identified a domain in the receptor that mediates ligand-independent assembly of the receptor.

    Article  CAS  PubMed  Google Scholar 

  43. Zhai, Y. et al. VEGI, a novel cytokine of the tumor necrosis factor family, is an angiogenesis inhibitor that suppresses the growth of colon carcinomas in vivo. FASEB J. 13, 181–189 (1999).

    Article  CAS  PubMed  Google Scholar 

  44. Schneider, P. et al. TRAIL receptors 1 (DR4) and 2 (DR5) signal FADD-dependent apoptosis and activate NF-κB. Immunity 7, 831–836 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Kischkel, F. C. et al. Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity 12, 611–620 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Ashkenazi, A. et al. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nature Rev. Cancer. 2, 420–430 (2002).

    Article  CAS  Google Scholar 

  47. Kwon, B. S. et al. TR1, a new member of the tumor necrosis factor receptor superfamily, induces fibroblast proliferation and inhibits osteoclastogenesis and bone resorption. FASEB J. 12, 845–854 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Grell, M. et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83, 793–802 (1995). This report shows that soluble TNF activates TNFR1 and membrane TNF activates TNFR2.

    Article  CAS  PubMed  Google Scholar 

  49. Weiss, T. et al. TNFR80-dependent enhancement of TNFR60-induced cell death is mediated by TNFR-associated factor 2 and is specific for TNFR60. J. Immunol. 161, 3136–3142 (1998).

    CAS  PubMed  Google Scholar 

  50. Mukhopadhyay, A. et al. Genetic deletion of the tumor necrosis factor receptor p60 or p80 abrogates ligand-mediated activation of nuclear factor-κB and of mitogen-activated protein kinases in macrophages. J. Biol. Chem. 276, 31906–31912 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Schneider, P. et al. TWEAK can induce cell death via endogenous TNF and TNF receptor 1. Eur. J. Immunol. 29, 1785–1792 (1999).

    Article  CAS  PubMed  Google Scholar 

  52. Grell, M. et al. Induction of cell death by tumour necrosis factor (TNF) receptor 2, CD40 and CD30: a role for TNF-R1 activation by endogenous membrane-anchored TNF. EMBO J. 18, 3034–3043 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Takada, Y. et al. Genetic deletion of the TNF receptor p60 or p80 sensitizes macrophages to lipopolysaccharide-induced nuclear factor-κB, mitogen-activated protein kinases and apoptosis. J. Biol. Chem. 278, 23390–23397 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Wajant, H. et al. Tumor necrosis factor signaling. Cell Death Differ. 10, 45–65 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Baud, V. et al. Signal transduction by tumor necrosis factor and its relatives. Trends Cell. Biol. 11, 372–377 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Chen, G. et al. TNF-R1 signaling: a beautiful pathway. Science 296, 1634–1635 (2002).

    Article  CAS  PubMed  Google Scholar 

  57. Harper, N. et al. Fas-associated death domain protein and caspase-8 are not recruited to the TNF-R1 signaling complex during TNF-induced apoptosis. J. Biol. Chem. 278, 25534–25541 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Nagata, S. et al. Fas ligand-induced apoptosis. Annu. Rev. Genet. 33, 29–55 (1999).

    Article  CAS  PubMed  Google Scholar 

  59. Pan, G. et al. The receptor for the cytotoxic ligand TRAIL. Science 276, 111–113 (1997).

    Article  CAS  PubMed  Google Scholar 

  60. Pan, G. et al. Identification and functional characterization of DR6, a novel death domain-containing TNF receptor. FEBS Lett. 431, 351–356 (1998).

    Article  CAS  PubMed  Google Scholar 

  61. Arch, R. H. et al. Tumor necrosis factor receptor-associated factors (TRAFs) — a family of adapter proteins that regulates life and death. Genes Dev. 12, 2821–2830 (1998).

    Article  CAS  PubMed  Google Scholar 

  62. Darnay, B. G. et al. Activation of NF-κB by RANK requires tumor necrosis factor receptor-associated factor (TRAF) 6 and NF-κB-inducing kinase. Identification of a novel TRAF6 interaction motif. J. Biol. Chem. 274, 7724–7731 (1999).

    Article  CAS  PubMed  Google Scholar 

  63. McWhirter, S. M. et al. Structural and biochemical analysis of signal transduction by the TRAF family of adapter proteins. Cold Spring Harb. Symp. Quant. Biol. 64, 551–562 (1999).

    Article  CAS  PubMed  Google Scholar 

  64. Li, X. et al. TNF-RII and c-IAP1 mediate ubiquitination and degradation of TRAF2. Nature 416, 345–347 (2002).

    Article  PubMed  Google Scholar 

  65. Claudio, E., Brown, K., Park, S., Wang, H. & Siebenlist, U. BAFF-induced NEMO-independent processing of NF-κB2 in maturing B cells. Nature Immunol. 3, 958–965 (2002).

    Article  CAS  Google Scholar 

  66. Kayagaki, N. et al. BAFF/BLyS receptor 3 binds the B cell survival factor BAFF ligand through a discrete surface loop and promotes processing of NF-κB2. Immunity 17, 515–524 (2002).

    Article  CAS  PubMed  Google Scholar 

  67. Dejardin, E. et al. The lymphotoxin-β receptor induces different patterns of gene expression via two NF-κB pathways. Immunity 17, 525–535 (2002).

    Article  CAS  PubMed  Google Scholar 

  68. Cao, Y. et al. IKKα provides an essential link between RANK signaling and cyclin D1 expression during mammary gland development. Cell 107, 763–775 (2001).

    Article  CAS  PubMed  Google Scholar 

  69. Legler, D. F., Micheau, O., Doucey, M. A., Tschopp, J. & Bron, C. Recruitment of TNF receptor 1 to lipid rafts is essential for TNFα-mediated NF-κB activation. Immunity 18, 655–664 (2003). This paper shows that after binding of TNF, TNFR1 translocates to cholesterol- and sphingolipid-enriched membrane microdomains, known as lipid rafts, in which it associates with receptor-interacting protein (RIP), TNFR-associated death domain (TRADD) and TNFR-associated factor 2 (TRAF2), forming a signalling complex.

    Article  CAS  PubMed  Google Scholar 

  70. Yeh, W. C. et al. Early lethality, functional NF-κB activation, and increased sensitivity to TNF-induced cell death in TRAF2-deficient mice. Immunity 7, 715–725 (1997).

    Article  CAS  PubMed  Google Scholar 

  71. Tada, K. et al. Critical roles of TRAF2 and TRAF5 in tumor necrosis factor-induced NF–κB activation and protection from cell death. J. Biol. Chem. 276, 36530–36534 (2001). Although a dominant-negative mutant of TRAF2 blocks TNF-induced nuclear factor-κB (NF-κB) activation, the deletion of TRAF2 has no marked effect on TNF-induced NF-κB activation. Here, the authors show that cells from TRAF2- and TRAF5-double-knockout mice are impaired in TNF-induced NF-κB activation.

    Article  CAS  PubMed  Google Scholar 

  72. Kelliher, M. A. et al. The death domain kinase RIP mediates the TNF-induced NF-κB signal. Immunity 8, 297–303 (1998).

    Article  CAS  PubMed  Google Scholar 

  73. Malinin, N. L., Boldin, M. P., Kovalenko, A. V. & Wallach, D. MAP3K-related kinase involved in NF-κB induction by TNF, CD95 and IL-1. Nature 385, 540–544 (1997).

    Article  CAS  PubMed  Google Scholar 

  74. Yin, L. et al. Defective lymphotoxin-βreceptor-induced NF-κB transcriptional activity in NIK-deficient mice. Science 291, 2162–2165 (2001).

    Article  CAS  PubMed  Google Scholar 

  75. Yang, J. et al. Mekk3 is essential for early embryonic cardiovascular development. Nature Genet. 24, 309–313 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Sugarman, B. J. et al. Recombinant human tumor necrosis factor-α: effects on proliferation of normal and transformed cells in vitro. Science 230, 943–945 (1985).

    Article  CAS  PubMed  Google Scholar 

  77. Karin, M. et al. NF-κB at the crossroads of life and death. Nature Immunol. 3, 221–227 (2002).

    Article  CAS  Google Scholar 

  78. Kasof, G. M. et al. Tumor necrosis factor-α induces the expression of DR6, a member of the TNF receptor family, through activation of NF-κB. Oncogene 20, 7965–7795 (2001).

    Article  CAS  PubMed  Google Scholar 

  79. Zheng, Y. et al. NF-κB RelA (p65) is essential for TNF-α-induced Fas expression but dispensable for both TCR-induced expression and activation-induced cell death. J. Immunol. 166, 4949–4957 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Ravi, R. et al. Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-κB. Nature Cell Biol. 3, 409–416 (2001).

    Article  CAS  PubMed  Google Scholar 

  81. You, Z. et al. c-Myc sensitizes cells to tumor necrosis factor-mediated apoptosis by inhibiting nuclear factor-κB transactivation. J. Biol. Chem. 277, 36671–36677 (2002).

    Article  CAS  PubMed  Google Scholar 

  82. Klefstrom, J. et al. Induction of TNF-sensitive cellular phenotype by c-Myc involves p53 and impaired NF-κB activation. EMBO J. 16, 7382–7392 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Feinberg, B. et al. A phase I trial of intravenously-administered recombinant tumor necrosis factor-α in cancer patients. J. Clin. Oncol. 6, 1328–1334 (1988).

    Article  CAS  PubMed  Google Scholar 

  84. Wielockx, B. et al. Inhibition of matrix metalloproteinases blocks lethal hepatitis and apoptosis induced by tumor necrosis factor and allows safe antitumor therapy. Nature Med. 7, 1202–1208 (2001).

    Article  CAS  PubMed  Google Scholar 

  85. Eggermont, A. M., de Wilt, J. H. & ten Hagen, T. L. Current uses of isolated limb perfusion in the clinic and a model system for new strategies. Lancet Oncol. 4, 429–437 (2003).

    Article  PubMed  Google Scholar 

  86. Lans, T. E. et al. Role of tumor necrosis factor on toxicity and cytokine production after isolated hepatic perfusion. Clin. Cancer Res. 7, 784–790 (2001).

    CAS  PubMed  Google Scholar 

  87. Kelley, S. K. et al. Preclinical studies to predict the disposition of Apo2L/tumor necrosis factor-related apoptosis-inducing ligand in humans: characterization of in vivo efficacy, pharmacokinetics, and safety. J. Pharmacol. Exp. Ther. 299, 31–38 (2001).

    CAS  PubMed  Google Scholar 

  88. Chuntharapai, A. et al. Isotype-dependent inhibition of tumor growth in vivo by monoclonal antibodies to death receptor 4. J. Immunol. 166, 4891–4898 (2001).

    Article  CAS  PubMed  Google Scholar 

  89. Ichikawa, K. et al. Tumoricidal activity of a novel anti-human DR5 monoclonal antibody without hepatocyte cytotoxicity. Nature Med. 7, 954–960 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Adams, A. B. et al. The role of TNF receptor and TNF superfamily molecules in organ transplantation. Am. J. Transplant. 2, 12–18 (2002).

    Article  CAS  PubMed  Google Scholar 

  91. Mckay, F. et al. BAFF AND APRIL: a tutorial on B cell survival. Annu. Rev. Immunol. 21, 231–264 (2003).

    Article  CAS  Google Scholar 

  92. Nagata, S. et al. The Fas death factor. Science 267, 1449–1456 (1995).

    Article  CAS  PubMed  Google Scholar 

  93. Kishimoto, H. et al. A role for Fas in negative selection of thymocytes in vivo. J. Exp. Med. 187, 1427–1438 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Hayashi, T. & Faustman, D. L. Role of defective apoptosis in type 1 diabetes and other autoimmune diseases. Recent Prog. Horm. Res. 58, 131–153 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Cohen, P. L. et al. Lpr and gld: single gene models of systemic autoimmunity and lymphoproliferative disease. Annu. Rev. Immunol. 9, 243–269 (1991).

    Article  CAS  PubMed  Google Scholar 

  96. Griffith, T. S. et al. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 270, 1189–1192 (1995).

    Article  CAS  PubMed  Google Scholar 

  97. Song, K. et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an inhibitor of autoimmune inflammation and cell cycle progression. J. Exp. Med. 191, 1095–1104 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Cretney, E. et al. Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice. J. Immunol. 168, 1356–1361 (2002).

    Article  CAS  PubMed  Google Scholar 

  99. Lamhamedi-Cherradi, S. E. et al. Defective thymocyte apoptosis and accelerated autoimmune diseases in TRAIL−/− mice. Nature Immunol. 4, 255–260 (2003).

    Article  CAS  Google Scholar 

  100. Gardam, M. A. et al. Anti-tumour necrosis factor agents and tuberculosis risk: mechanisms of action and clinical management. Lancet Infect. Dis. 3, 148–155 (2003).

    Article  CAS  PubMed  Google Scholar 

  101. Moore, R. J. et al. Mice deficient in tumor necrosis factor-α are resistant to skin carcinogenesis. Nature Med. 5, 828–831 (1999). Given the associations between chronic inflammation and epithelial cell cancer, the authors studied susceptibility to skin carcinogenesis in mice deficient for TNF. TNF−/− mice were resistant to the development of benign and malignant skin tumours, whether induced by initiation with DMBA (12-dimethylbenz[a]-anthracene) and promotion with TPA (12-0-tetradecanoyl-phorbol-13-acetate) or by repeated dosing with DMBA.

    Article  CAS  PubMed  Google Scholar 

  102. Dajee, M. et al. NF-κB blockade and oncogenic Ras trigger invasive human epidermal neoplasia. Nature 421, 639–643 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Hotamisligil, G. S. et al. Free in PMC tumor necrosis factor-α inhibits signaling from the insulin receptor. Proc. Natl Acad. Sci. USA 91, 4854–4858 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Emanuelli, B. et al. SOCS-3 inhibits insulin signaling and is upregulated in response to tumor necrosis factor-α in the adipose tissue of obese mice. J. Biol. Chem. 276, 47944–47949 (2001).

    Article  CAS  PubMed  Google Scholar 

  105. Uysal, K. T. et al. Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature 389, 610–614 (1997). The authors investigated the role of TNF in obesity and insulin resistance, by generating obese mice with a targeted null mutation in the gene encoding TNF and those encoding the two receptors for TNF. The absence of TNF improved insulin sensitivity in both diet-induced obesity and that resulting for the ob/ob model of obesity.

    Article  CAS  PubMed  Google Scholar 

  106. Bolger, A. P. et al. Tumour necrosis factor in chronic heart failure: a peripheral view on pathogenesis, clinical manifestations and therapeutic implications. Drugs 60, 1245–1257 (2000).

    Article  CAS  PubMed  Google Scholar 

  107. Diwan, A. et al. Inflammatory mediators and the failing heart: a translational approach. Curr. Mol. Med. 3, 161–182 (2003).

    Article  CAS  PubMed  Google Scholar 

  108. Romas, E. et al. Involvement of receptor activator of NF-κB ligand and tumor necrosis factor-α in bone destruction in rheumatoid arthritis. Bone 30, 340–346 (2002).

    Article  CAS  PubMed  Google Scholar 

  109. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315–323 (1999). Bone remodelling and bone loss are controlled by a balance between osteoprotegerin ligand (OPGL) and OPG. In addition, OPGL regulates lymph-node organogenesis, lymphocyte development and interactions between T cells and dendritic cells. Here, the authors report that activated T cells can directly trigger osteoclastogenesis through the expression of OPGL.

    Article  CAS  PubMed  Google Scholar 

  110. Fata, J. E. et al. The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell 103, 41–50 (2000).

    Article  CAS  PubMed  Google Scholar 

  111. Bucay, N. et al. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev. 12, 1260–1268 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Aganna, E. et al. Tumor necrosis factor receptor-associated periodic syndrome (TRAPS) in a Dutch family: evidence for a TNFRSF1A mutation with reduced penetrance. Eur. J. Hum. Genet. 9, 63–66 (2001).

    Article  CAS  PubMed  Google Scholar 

  113. Nagata, S. et al. Human autoimmune lymphoproliferative syndrome, a defect in the apoptosis-inducing Fas receptor: a lesson from the mouse model. J. Hum. Genet. 43, 2–8 (1998).

    Article  CAS  PubMed  Google Scholar 

  114. Straus, S. E. et al. The development of lymphomas in families with autoimmune lymphoproliferative syndrome with germline Fas mutations and defective lymphocyte apoptosis. Blood 98, 194–200 (2001).

    Article  CAS  PubMed  Google Scholar 

  115. Chun, H. J. et al. Pleiotropic defects in lymphocyte activation caused by caspase-8 mutations lead to human immunodeficiency. Nature 419, 395–399 (2002).

    Article  CAS  PubMed  Google Scholar 

  116. Ramesh, N. et al. CD40–CD40 ligand (CD40L) interactions and X-linked hyperIgM syndrome (HIGMX–1). Clin. Immunol. Immunopathol. 76, S208–S213 (1995).

    Article  CAS  PubMed  Google Scholar 

  117. Hughes, A. E. et al. Mutations in TNFRSF11A, affecting the signal peptide of RANK, cause familial expansile osteolysis. Nature Genet. 24, 45–48 (2000).

    Article  CAS  PubMed  Google Scholar 

  118. Thesleff, I. & Mikkola, M. L. Death receptor signaling giving life to ectodermal organs. Sci. STKE 2002, PE22 (2002).

    PubMed  Google Scholar 

  119. Takakuwa, T. et al. Frequent mutations of Fas gene in thyroid lymphoma. Cancer Res. 61, 1382–1385 (2001).

    CAS  PubMed  Google Scholar 

  120. Takayama, H. et al. Fas gene mutations in prostatic intraepithelial neoplasia and concurrent carcinoma: analysis of laser capture microdissected specimens. Lab. Invest. 81, 283–288 (2001).

    Article  CAS  PubMed  Google Scholar 

  121. Takayama, H. et al. Frequent Fas gene mutations in testicular germ cell tumors. Am. J. Pathol. 161, 635–641 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Fisher, M. J. et al. Nucleotide substitution in the ectodomain of trail receptor DR4 is associated with lung cancer and head and neck cancer. Clin. Cancer Res. 7, 1688–1697 (2001).

    CAS  PubMed  Google Scholar 

  123. Suenaert, P. et al. Anti-tumor necrosis factor treatment restores the gut barrier in Crohn's disease. Am. J. Gastroenterol. 97, 2000–2004 (2002).

    Article  CAS  PubMed  Google Scholar 

  124. Couriel, D. R. et al. Role of tumor necrosis factor-α inhibition with inflixiMAB in cancer therapy and hematopoietic stem cell transplantation. Curr. Opin. Oncol. 12, 582–587 (2000).

    Article  CAS  PubMed  Google Scholar 

  125. Feldmann, M. et al. Anti-TNFα therapy of rheumatoid arthritis: what have we learned? Annu. Rev. Immunol. 19, 163–196 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by The Clayton Foundation for Research, by the Department of Defense of the US Army Breast Cancer Research Program and by the National Institutes of Health. I would like to thank Y. Takeda, A. Bharti, U. Bhardwaj and S. Shishodia for assistance with the graphics, U. Gaur and L. Ford for help in preparation of the manuscript and W. Pagel for a careful review of the manuscript.

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASES

LocusLink

4-1BB

APRIL

BAFF

BAFFR

BCMA

CD27

CD30

CD30L

CD40

CD40L

CD95

CD95L

DCR1

DCR2

DCR3

DR4

DR5

DR6

EDA

EDAR

GITRL

HVEM

IAP1

IKKα

IKKβ

IKKγ

LT

LTβR

MEKK3

OPG

OX40

RANKL

RELT

RIP

SOCS3

TACI

TNF

TNFR1

TNFR2

TRADD

TRAF1

TRAF2

TRAF5

TRAF6

TRAIL

TRAILR1

TROY

TWEAK

VEGI

FURTHER INFORMATION

Bharat Aggarwal's lab

Glossary

SYSTEMIC LUPUS ERYTHEMATOSUS

(SLE). A disease of unknown origin in which tissues and cells are damaged by the deposition of pathogenic antibodies and immune complexes. Patients generally have abnormal B- and T-cell functions.

SJOGREN'S SYNDROME

(SS). An autoimmune disease characterized by diffuse lymphoid-cell infiltrates in the salivary and lacrimal glands, resulting in dry eyes and mouth due to insufficient secretion.

GRAFT-VERSUS-HOST DISEASE

(GVHD). Tissue damage in a recipient of allogeneic transplanted tissue (usually a bone-marrow transplant) that results from the activity of donor cytotoxic T cells that recognize the recipient's tissue as foreign. GVHD varies markedly in severity, but can be life threatening in severe cases. Typically, damage to the skin and gut mucosa leads to clinical manifestations.

IMMUNE-PRIVILEGED SITE

Immune-privileged sites are areas in the body with a decreased immune response to foreign antigens, including tissue grafts. These sites include the brain, eye, testis and uterus.

OSTEOPETROSIS

A hereditary bone disease with intense positive balance of body calcium. Autosomal recessive osteopetrosis is a rare, fatal disease characterized by the accumulation of excessive bone mass due to defective bone resorption. The pathogenesis of osteopetrosis is controversial. Defects in osteoblast–osteoclast interactions, incorrect differentiation of osteoclasts, abnormal contact between osteoclasts and the extracellular matrix, and abolished signalling can occur in this disease.

OSTEOPOROSIS

A condition that involves loss of bone due to an increase in the number of osteoclasts.

CANALE-SMITH SYNDROME

(CSS). An inherited disease characterized by massive lymphadenopathy, hepatosplenomegaly and systemic autoimmunity to erythrocytes and platelets.

AUTOIMMUNE LYMPHOPROLIFERATIVE SYNDROME

(ALPS). ALPS is characterized clinically by chronic non-malignant lymphoproliferation and autoimmunity, and is caused by a genetic defect in apoptosis. Most patients with ALPS have heterozygous mutations in the CD95 gene.

OSTEOBLASTS

Cells that are responsible for the formation of bone.

OSTEOCLASTS

Cells that are responsible for bone resorption. They are rare cells with only 2–3 cells seen per 1 mm3 of bone. However, the loss of function in osteoclasts, problems with their differentiation and decrease in their number lead to bone osteosclerosis/osteopetrosis. Conversely, an increase in their number or function induces bone osteoporosis, indicating that osteoclasts have a pivotal role in bone homeostasis.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Aggarwal, B. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol 3, 745–756 (2003). https://doi.org/10.1038/nri1184

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri1184

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing