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:

Forkhead-box transcription factors and their role in the immune system

Key Points

  • More than 100 forkhead transcription factors have been identified, and they are known as FOX (forkhead box) proteins. These proteins are classified in terms of structure, not function, and mutation of various FOX proteins leads to immune dysfunction.

  • FOXP3 regulates the production of CD4+CD25+ regulatory T cells, which are crucial for the maintenance of self-tolerance. Deletion of the gene encoding FOXP3 results in overproliferation of activated T cells and development of both autoimmune disease and allergy.

  • FOXN1 is crucial for the growth and differentiation of thymic epithelial cells. Deletion or mutation of the gene encoding this transcription factor, as found in the nude mouse, results in athymia.

  • FOXJ1 is a transcriptional activator, and its expression levels are downregulated by lymphocytes from mice prone to systemic lupus erythematosus. FOXJ1 regulates T-cell activation, probably through inhibiting the activation of nuclear factor-κB (NF-κB).

  • FOXO-subfamily members have central roles in the regulation of proliferation and apoptosis of various cell types. In the immune system, FOXO activity tends to be inversely correlated with cellular activation.

  • Deletion of the gene encoding FOXO3A results in lymphoproliferation and multi-organ inflammation. Similar to FOXJ1, this might also occur through modulation of NF-κB activity.

Abstract

It is more than a decade since the discovery of the first forkhead-box (FOX) transcription factor in the fruit fly Drosophila melanogaster. In the intervening time, there has been an explosion in the identification and characterization of members of this family of proteins. Importantly, in the past few years, it has become clear that members of the FOX family have crucial roles in various aspects of immune regulation, from lymphocyte survival to thymic development. This review focuses on FOXP3, FOXN1, FOXJ1 and members of the FOXO subfamily and their function in the immune system.

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

Access options

Buy this article

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

Figure 1: The FOX protein winged-helix DNA-binding domain.
Figure 2: FOXP3 is a master regulator of regulatory T cells.
Figure 3: Thymic development and FOXN1.
Figure 4: Regulation of TH1-cell activation by FOXJ1 and FOXO3A.
Figure 5: Regulation and function of FOXO transcription factors.

Similar content being viewed by others

References

  1. Kaufmann, E. & Knochel, W. Five years on the wings of fork head. Mech. Dev. 57, 3–20 (1996).

    Article  CAS  Google Scholar 

  2. Weigel, D., Jurgens, G., Kuttner, F., Seifert, E. & Jackle, H. The homeotic gene fork head encodes a nuclear protein and is expressed in the terminal regions of the Drosophila embryo. Cell 57, 645–658 (1989).

    Article  CAS  Google Scholar 

  3. Lai, E., Prezioso, V. R., Smith, E., Litvin, O., Costa, R. H. & Darnell, J. E. HNF-3A, a hepatocyte-enriched transcription factor of novel structure is regulated transcriptionally. Genes Dev. 4, 1427–1436 (1990).

    Article  CAS  Google Scholar 

  4. Kaestner, K. H., Knochel, W. & Martinez, D. E. Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev. 14, 142–146 (2000).

    CAS  PubMed  Google Scholar 

  5. Clark, K. L., Halay, E. D., Lai, E. & Burley, S. K. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 364, 412–420 (1993).

    Article  CAS  Google Scholar 

  6. Lehmann, O. J., Sowden, J. C., Carlsson, P., Jordan, T. & Bhattacharya, S. S. Fox's in development and disease. Trends Genet. 19, 339–344 (2003).

    Article  CAS  Google Scholar 

  7. Maloy, K. J. & Powrie, F. Regulatory T cells in the control of immune pathology. Nature Immunol. 2, 816–822 (2001).

    Article  CAS  Google Scholar 

  8. Shevach, E. M. CD4+CD25+ suppressor T cells: more questions than answers Nature Rev. Immunol. 2, 389–400 (2002).

    Article  CAS  Google Scholar 

  9. Blair, P. J. et al. CD4+CD8 T cells are the effector cells in disease pathogenesis in the scurfy (sf) mouse. J. Immunol. 153, 3764–3774 (1994).

    CAS  PubMed  Google Scholar 

  10. Godfrey, V. L., Wilkinson, J. E., Rinchik, E. M. & Russell, L. B. Fatal lymphoreticular disease in the scurfy (sf) mouse requires T cells that mature in a sf thymic environment: potential model for thymic education. Proc. Natl Acad. Sci. USA 88, 5528–5532 (1991).

    Article  CAS  Google Scholar 

  11. Brunkow, M. E. et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nature Genet. 27, 68–73 (2001). This study identified the gene that is defective in scurfy mice by combining high-resolution genetic and physical mapping with large-scale sequence analysis. Genetic complementation showed that the protein product of Foxp3 is essential for normal immune homeostasis.

    Article  CAS  Google Scholar 

  12. Ramsdell, F. Foxp3 and natural regulatory T cells: key to a cell lineage? Immunity 19, 165–168 (2003).

    Article  CAS  Google Scholar 

  13. Wildin, R. S. et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nature Genet. 27, 18–20 (2001).

    Article  CAS  Google Scholar 

  14. Bennett, C. L. et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nature Genet. 27, 20–21 (2001).

    Article  CAS  Google Scholar 

  15. Chatila, T. A. et al. JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J. Clin. Invest. 106, R75–R81 (2000).

    Article  CAS  Google Scholar 

  16. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunol. 4, 330–336 (2003).

    Article  CAS  Google Scholar 

  17. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science 299, 1057–1061 (2003). References 16 and 17 showed that FOXP3 is specifically expressed by CD4+CD25+ T Reg cells and is required for their development. Furthermore, expression of FOXP3 was found to confer suppressor function on peripheral CD4+CD25 T cells. In addition, reference 17 showed that FOXP3, which encodes a transcription factor that is genetically defective in an autoimmune and inflammatory syndrome in humans and mice, is specifically expressed by naturally arising CD4+ regulatory T cells.

    Article  CAS  Google Scholar 

  18. Khattri, R. et al. The amount of scurfin protein determines peripheral T cell number and responsiveness. J. Immunol. 167, 6312–6320 (2001).

    Article  CAS  Google Scholar 

  19. Asseman, C., Mauze, S., Leach, M. W., Coffman, R. L. & Powrie, F. An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. J. Exp. Med. 190, 995–1004 (1999).

    Article  CAS  Google Scholar 

  20. Schubert, L. A., Jeffery, E., Zhang, Y., Ramsdell, F. & Ziegler, S. F. Scurfin (FOXP3) acts as a repressor of transcription and regulates T cell activation. J. Biol. Chem. 276, 37672–37679 (2001).

    Article  CAS  Google Scholar 

  21. Walker, M. R. et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25 T cells. J. Clin. Invest. 112, 1437–1443 (2003).

    Article  CAS  Google Scholar 

  22. Baecher-Allan, C., Brown, J. A., Freeman, G. J. & Hafler, D. A. CD4+CD25hi regulatory cells in human peripheral blood. J. Immunol. 167, 1245–1253 (2001).

    Article  CAS  Google Scholar 

  23. Nakamura, K., Kitani, A. & Strober, W. Cell contact-dependent immunosuppression by CD4+CD25+ regulatory T cells is mediated by cell surface-bound transforming growth factor β. J. Exp. Med. 194, 629–644 (2001).

    Article  CAS  Google Scholar 

  24. Chen, W. et al. Conversion of peripheral CD4+CD25 naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3. J. Exp. Med. 198, 1875–1886 (2003).

    Article  CAS  Google Scholar 

  25. Fantini, M. C. et al. TGF-β induces a regulatory phenotype in CD4+CD25 T cells through Foxp3 induction and down-regulation of Smad7. J. Immunol. 172, 5149–5153 (2004). This study showed that TGF-β induces a regulatory phenotype in CD4+CD25 T cells through the induction of FOXP3 expression, which can subsequently inhibit SMAD7 and thereby activate a positive autoregulatory loop of TGF-β signalling.

    Article  CAS  Google Scholar 

  26. Peng, Y., Laouar, Y., Li, M. O., Green, E. A. & Flavell, R. A. TGF-β regulates in vivo expansion of Foxp3-expressing CD4+CD25+ regulatory T cells responsible for protection against diabetes. Proc. Natl Acad. Sci. USA 101, 4572–4577 (2004).

    Article  CAS  Google Scholar 

  27. Mamura, M. et al. CD28 disruption exacerbates inflammation in Tgf-β1−/− mice: in vivo suppression by CD4+CD25+ regulatory T cells independent of autocrine TGF-β1. Blood 103, 4594–4601 (2004).

    Article  CAS  Google Scholar 

  28. Polanczyk, M. J. et al. Estrogen drives expansion of the CD4+CD25+ regulatory T cell compartment. J. Immunol. 173, 2227–2230 (2004).

    Article  CAS  Google Scholar 

  29. Blackburn, C. C. & Manley, N. R. Developing a new paradigm for thymus organogenesis. Nature Rev. Immunol. 4, 278–289 (2004).

    Article  CAS  Google Scholar 

  30. Nehls, M., Pfeifer, D., Schorpp, M., Hedrich, H. & Boehm, T. New member of the winged-helix protein family disrupted in mouse and rat nude mutations. Nature 372, 103–107 (1994).

    Article  CAS  Google Scholar 

  31. Bleul, C. C. & Boehm, T. Laser capture microdissection-based expression profiling identifies PD1-ligand as a target of the nude locus gene product. Eur. J. Immunol. 31, 2497–2503 (2001).

    Article  CAS  Google Scholar 

  32. Nishimura, H., Honjo, T. & Minato, N. Facilitation of β selection and modification of positive selection in the thymus of PD-1-deficient mice. J. Exp. Med. 191, 891–898 (2000).

    Article  CAS  Google Scholar 

  33. Balciunaite, G. et al. Wnt glycoproteins regulate the expression of FoxN1, the gene defective in nude mice. Nature Immunol. 3, 1102–1108 (2002). This paper reported that secreted WNT glycoproteins expressed by TECs and thymocytes can regulate FOXN1 expression in the epithelium through both autocrine and paracrine mechanisms.

    Article  CAS  Google Scholar 

  34. Tsai, P. T., Lee, R. A. & Wu, H. BMP4 acts upstream of FGF in modulating thymic stroma and regulating thymopoiesis. Blood 102, 3947–3953 (2003).

    Article  CAS  Google Scholar 

  35. Staal, F. J. & Clevers, H. C. Wnt signaling in the thymus. Curr. Opin. Immunol. 15, 204–208 (2003).

    Article  CAS  Google Scholar 

  36. Baxter, R. M. & Brissette, J. L. Role of the nude gene in epithelial terminal differentiation. J. Invest. Dermatol. 118, 303–309 (2002).

    Article  CAS  Google Scholar 

  37. Su, D. M., Navarre, S., Oh, W. J., Condie, B. G. & Manley, N. R. A domain of Foxn1 required for crosstalk-dependent thymic epithelial cell differentiation. Nature Immunol. 4, 1128–1135 (2003).

    Article  CAS  Google Scholar 

  38. Schlake, T., Schorpp, M. and Boehm, T. Formation of regulator/target gene relationships during evolution. Gene 256, 29–34 (2000).

    Article  CAS  Google Scholar 

  39. Hackett, B. P. et al. Primary structure of hepatocyte nuclear factor/forkhead homologue 4 and characterization of gene expression in the developing respiratory and reproductive epithelium. Proc. Natl Acad. Sci. USA 92, 4249–4253 (1995).

    Article  CAS  Google Scholar 

  40. Clevidence, D. E. et al. Members of the HNF-3/forkhead family of transcription factors exhibit distinct cellular expression patterns in lung and regulate the surfactant protein B promoter. Dev. Biol. 166, 195–209 (1994).

    Article  CAS  Google Scholar 

  41. Lim, L., Zhou, H. & Costa, R. H. The winged helix transcription factor HFH-4 is expressed during choroid plexus epithelial development in the mouse embryo. Proc. Natl Acad. Sci. USA 94, 3094–3099 (1997).

    Article  CAS  Google Scholar 

  42. Chen, J., Knowles, H. J., Hebert, J. L. & Hackett, B. P. Mutation of the mouse hepatocyte nuclear factor/forkhead homologue 4 gene results in an absence of cilia and random left–right asymmetry. J. Clin. Invest. 102, 1077–1082 (1998).

    Article  CAS  Google Scholar 

  43. Brody, S. L., Yan, X. H., Wuerffel, M. K., Song, S. K. & Shapiro, S. D. Ciliogenesis and left–right axis defects in forkhead factor HFH-4-null mice. Am. J. Respir. Cell. Mol. Biol. 23, 45–51 (2000).

    Article  CAS  Google Scholar 

  44. Lin, L., Spoor, M. S., Gerth, A. J., Brody, S. L. & Peng, S. L. Modulation of TH1 activation and inflammation by the NF-κB repressor Foxj1. Science 303, 1017–1020 (2004). This study showed that FOXJ1 can inhibit NF-κB signalling through induction of IκB proteins. These results indicate that FOXJ1 might modulate inflammatory reactions and prevent autoimmunity by antagonizing the transcription of genes that encode pro-inflammatory cytokines.

    Article  CAS  Google Scholar 

  45. Rao, A., Luo, C. & Hogan, P. G. Transcription factors of the NFAT family: regulation and function. Annu. Rev. Immunol. 15, 707–747 (1997).

    Article  CAS  Google Scholar 

  46. Li, Q. & Verma, I. M. NF-κB regulation in the immune system. Nature Rev. Immunol. 2, 725–734 (2002).

    Article  CAS  Google Scholar 

  47. Kojima, H. et al. An essential role for NF-κB in IL-18-induced IFN-γ expression in KG-1 cells. J. Immunol. 162, 5063–5069 (1999).

    CAS  PubMed  Google Scholar 

  48. Brody, S. L., Hackett, B. P. & White, R. A. Structural characterization of the mouse Hfh4 gene, a developmentally regulated forkhead family member. Genomics 45, 509–518 (1997).

    Article  CAS  Google Scholar 

  49. Schade, A. E. & Levine, A. D. Extracellular signal-regulated kinases 1/2 function as integrators of TCR signal strength. J. Immunol. 172, 5828–5832 (2004).

    Article  CAS  Google Scholar 

  50. Jorritsma, P. J., Brogdon, J. L. & Bottomly, K. Role of TCR-induced extracellular signal-regulated kinase activation in the regulation of early IL-4 expression in naive CD4+ T cells. J. Immunol. 170, 2427–2434 (2003).

    Article  CAS  Google Scholar 

  51. Burgering, B. M. & Medema, R. H. Decisions on life and death: FOXO forkhead transcription factors are in command when PKB/Akt is off duty. J. Leukoc. Biol. 73, 689–701 (2003).

    Article  CAS  Google Scholar 

  52. Birkenkamp, K. U. & Coffer, P. J. FOXO transcription factors as regulators of immune homeostasis: molecules to die for? J. Immunol. 171, 1623–1629 (2003).

    Article  CAS  Google Scholar 

  53. Accili, D. & Arden, K. C. FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 117, 421–426 (2004).

    Article  CAS  Google Scholar 

  54. Lin, L., Hron, J. D. & Peng, S. L. Regulation of NF-κB, TH activation, and autoinflammation by the forkhead transcription factor Foxo3a. Immunity 21, 203–213 (2004). This was the first study that identified a role for FOXO transcription factors in immune homeostasis in vivo . FOXO3A can inhibit NF-κB activation and maintain T-cell tolerance through an undefined mechanism.

    Article  CAS  Google Scholar 

  55. Castrillon, D. H., Miao, L., Kollipara, R., Horner, J. W. & DePinho, R. A. Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science 301, 215–218 (2003).

    Article  CAS  Google Scholar 

  56. Medema, R. H., Kops, G. J., Bos, J. L. & Burgering, B. M. AFX-like forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1. Nature 404, 782–787 (2000). This paper showed that FOXO transcription factors can directly modulate proliferation.

    Article  CAS  Google Scholar 

  57. Kops, G. J. et al. Control of cell cycle exit and entry by protein kinase B-regulated forkhead transcription factors. Mol. Cell. Biol. 22, 2025–2036 (2002).

    Article  CAS  Google Scholar 

  58. Dijkers, P. F. et al. Forkhead transcription factor FKHR-L1 modulates cytokine-dependent transcriptional regulation of p27KIP1. Mol. Cell. Biol. 20, 9138–9148 (2000).

    Article  CAS  Google Scholar 

  59. Kops, G. J. et al. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419, 316–321 (2002).

    Article  CAS  Google Scholar 

  60. Schwartz, R. H. T cell anergy. Annu. Rev. Immunol. 21, 305–334 (2003).

    Article  CAS  Google Scholar 

  61. Jackson, S. K., DeLoose, A. & Gilbert, K. M. Induction of anergy in TH1 cells associated with increased levels of cyclin-dependent kinase inhibitors p21Cip1 and p27Kip1. J. Immunol. 166, 952–958 (2001).

    Article  CAS  Google Scholar 

  62. Stahl, M. et al. The forkhead transcription factor FoxO regulates transcription of p27Kip1 and Bim in response to IL-2. J. Immunol. 168, 5024–5031 (2002).

    Article  CAS  Google Scholar 

  63. Appleman, L. J., van Puijenbroek, A. A., Shu, K. M., Nadler, L. M. & Boussiotis, V. A. CD28 costimulation mediates down-regulation of p27kip1 and cell cycle progression by activation of the PI3K/PKB signaling pathway in primary human T cells. J. Immunol. 168, 2729–2736 (2002).

    Article  CAS  Google Scholar 

  64. Okkenhaug, K., Bilancio, A., Emery, J. L. & Vanhaesebroeck, B. Phosphoinositide 3-kinase in T cell activation and survival. Biochem. Soc. Trans. 32, 332–335 (2004).

    Article  CAS  Google Scholar 

  65. Martinez-Gac, L., Marques, M., Garcia, Z., Campanero, M. R. & Carrera, A. C. Control of cyclin G2 mRNA expression by forkhead transcription factors: mechanism for cell cycle control by phosphoinositide 3-kinase and forkhead. Mol. Cell. Biol. 24, 2181–2189 (2004).

    Article  CAS  Google Scholar 

  66. Horne, M. C. et al. Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated cell cycle arrest. J. Biol. Chem. 272, 12650–12661 (1997).

    Article  CAS  Google Scholar 

  67. Dijkers, P. F. et al. FKHR-L1 can act as a critical effector of cell death induced by cytokine withdrawal: protein kinase B-enhanced cell survival through maintenance of mitochondrial integrity. J. Cell Biol. 156, 531–542 (2002). This paper showed that activation of FOXO3A alone can recapitulate all known elements of the apoptotic programme that are normally induced in lymphocytes by cytokine withdrawal.

    Article  CAS  Google Scholar 

  68. Pandiyan, P. et al. CD152 (CTLA-4) determines the unequal resistance of TH1 and TH2 cells against activation-induced cell death by a mechanism requiring PI3 kinase function. J. Exp. Med. 199, 831–842 (2004).

    Article  CAS  Google Scholar 

  69. Jones, R. G. et al. Protein kinase B regulates T lymphocyte survival, nuclear factor κB activation, and Bcl-XL levels in vivo. J. Exp. Med. 191, 1721–1734 (2000).

    Article  CAS  Google Scholar 

  70. Tang, T. T. et al. The forkhead transcription factor AFX activates apoptosis by induction of the BCL-6 transcriptional repressor. J. Biol. Chem. 277, 14255–14265 (2002).

    Article  CAS  Google Scholar 

  71. Hu, M. C. et al. IκB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 117, 225–237 (2004).

    Article  CAS  Google Scholar 

  72. Pasparakis, M., Schmidt-Supprian, M. & Rajewsky, K. IκB kinase signaling is essential for maintenance of mature B cells. J. Exp. Med. 196, 743–752 (2002).

    Article  CAS  Google Scholar 

  73. Yusuf, I., Zhu, X., Kharas, M. G., Chen, J. & Fruman, D. A. Optimal B-cell proliferation requires phosphoinositide 3-kinase-dependent inactivation of FOXO transcription factor. Blood 104, 784–787 (2004).

    Article  CAS  Google Scholar 

  74. Bakker, W. J. et al. FoxO3a regulates erythroid differentiation and induces BTG1, an activator of protein arginine methyl transferase 1. J. Cell Biol. 164, 175–184 (2004).

    Article  CAS  Google Scholar 

  75. Frank, J. et al. Exposing the human nude phenotype. Nature 398, 473–474 (1999).

    Article  CAS  Google Scholar 

  76. Scheijen, B., Ngo, H. T., Kang, H. & Griffin, J. D. FLT3 receptors with internal tandem duplications promote cell viability and proliferation by signaling through Foxo proteins. Oncogene 23, 3338–3349 (2004).

    Article  CAS  Google Scholar 

  77. Komatsu, N. et al. A member of Forkhead transcription factor FKHRL1 is a downstream effector of STI571-induced cell cycle arrest in BCR-ABL-expressing cells. J. Biol. Chem. 278, 6411–6419 (2003).

    Article  CAS  Google Scholar 

  78. Ramaswamy, S., Nakamura, N., Sansal, I., Bergeron, L. & Sellers, W. R. A novel mechanism of gene regulation and tumor suppression by the transcription factor FKHR. Cancer Cell 2, 81–91 (2002).

    Article  CAS  Google Scholar 

  79. Carlsson, P. & Mahlapuu, M. Forkhead transcription factors: key players in development and metabolism. Dev. Biol. 250, 1–23 (2002).

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Paul J. Coffer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

Entrez Gene

FOXJ1

FOXN1

FOXO3A

FOXP3

IKK

PKB

PI3K

SMAD7

TGF-β

FURTHER INFORMATION

Paul Coffer's laboratory

Boudewijn Burgering's laboratory

Glossary

REGULATORY T (TReg) CELLS

A subset of CD4+ suppressor T cells that express high levels of CD25 (the interleukin-2 receptor α-chain), the role of which is to maintain self-tolerance.

RECOMBINATION-ACTIVATING-GENE-KNOCKOUT MICE

Recombination-activating genes (Rag1 and Rag2) are expressed by developing lymphocytes. Mice that are deficient in either RAG protein fail to produce B and T cells owing to a developmental block in the gene rearrangement that is required for receptor expression.

ARREST

Any process by which progression through the cell cycle is halted during one of the normal phases — G1 (gap 1), S (synthesis), G2 (gap 2) or M (mitosis).

WNT PROTEINS

WNTs are glycoproteins related to the Drosophila melanogaster protein Wingless, a ligand that regulates the temporal and spatial development of the embryo. WNT-mediated signalling has been shown to regulate cell-fate determination, proliferation, adhesion, migration and polarity during development. In addition to their crucial role in embryogenesis, WNTs and their downstream signalling molecules have been implicated in tumorigenesis and have causative roles in human colon cancers.

BONE MORPHOGENETIC PROTEINS

(BMPs). The genes encoding BMPs constitute a subfamily of the transforming growth factor-β gene superfamily. BMPs have a crucial role in the modulation of mesenchymal differentiation and the induction of cartilage and bone formation.

ANERGY

A state of T cells that have been stimulated through their T-cell receptors in the absence of ligation of CD28. On restimulation, these T cells are unable to produce interleukin-2 or to proliferate, even in the presence of co-stimulatory signals.

PROGRAMMED CELL DEATH

A common form of cell death, which is also known as apoptosis. Many physiological and developmental stimuli cause apoptosis, and this mechanism is frequently used to delete unwanted, superfluous or potentially harmful cells, such as those undergoing transformation. Apoptosis involves cell shrinkage, chromatin condensation in the periphery of the nucleus, plasma-membrane blebbing and DNA fragmentation into segments of about 180 base pairs. Eventually, the cell breaks up into many membrane-bound 'apoptotic bodies', which are phagocytosed by neighbouring cells.

HYPERMORPHIC

A type of mutation in which the altered gene product has an increased level of activity or in which the wild-type gene product is expressed at an increased level.

BCR-ABL

A tyrosine-kinase oncogene. The Abelson leukaemia-virus protein (ABL) is fused with the breakpoint-cluster region (BCR) in the Philadelphia-chromosome translocation found in chronic myeloid leukaemia.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Coffer, P., Burgering, B. Forkhead-box transcription factors and their role in the immune system. Nat Rev Immunol 4, 889–899 (2004). https://doi.org/10.1038/nri1488

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri1488

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