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:

Notch signalling: a simple pathway becomes complex

Key Points

  • Notch is the receptor in a highly conserved signalling pathway that is crucial in development and implicated in malignant transformation. The basic paradigm of Notch signalling is simple, and involves proteolytic cleavage to release an intracellular fragment (Nicd) that functions to regulate transcription.

  • In the nucleus, Nicd displaces a repression complex and, with the DNA-binding CSL (CBF1, Su(H) and LAG-1) protein, recruits the co-activator Mastermind. Additional epigenetic cofactors are implicated and recruitment of kinases and ubiquitin ligases probably contribute to rapid turnover of the activator complex.

  • Activity of the receptor is also regulated post-translationally. A number of different auxiliary components are implicated, including several ubiquitin ligases and proteins, such as Numb, that have direct links to the endocytic machinery.

  • Notch ligands are transmembrane proteins and they require E3 ubiquitin ligases for their activity. The mechanisms whereby the ligands become competent to signal are not yet known, but probably entail endocytosis. Localization and cleavage of ligands might also contribute to their regulation.

  • In many developmental processes, feedback mechanisms operate to amplify or restrict the activity of the Notch pathway. These include transcriptional regulation of auxiliary factors that in turn influence ligand or receptor activity and microRNAs.

Abstract

A small number of signalling pathways are used iteratively to regulate cell fates, cell proliferation and cell death in development. Notch is the receptor in one such pathway, and is unusual in that most of its ligands are also transmembrane proteins; therefore signalling is restricted to neighbouring cells. Although the intracellular transduction of the Notch signal is remarkably simple, with no secondary messengers, this pathway functions in an enormous diversity of developmental processes and its dysfunction is implicated in many cancers.

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: The core Notch pathway.
Figure 2: Ligand activation entails ubiquitylation.
Figure 3: Processing and trafficking regulate Notch-receptor activity.
Figure 4: Nuclear cycle of CSL.

Similar content being viewed by others

References

  1. Artavanis-Tsakonas, S., Rand, M. D. & Lake, R. J. Notch signaling: cell fate control and signal integration in development. Science 284, 770–776 (1999). This is an excellent review of the field prior to 1999.

    CAS  PubMed  Google Scholar 

  2. Schweisguth, F. Notch signaling activity. Curr. Biol. 14, R129–R138 (2004).

    CAS  PubMed  Google Scholar 

  3. Radtke, F. & Raj, K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nature Rev. Cancer 3, 756–767 (2003). A review that summarizes the links between Notch and cancer.

    CAS  Google Scholar 

  4. Fortini, M. E. γ-secretase-mediated proteolysis in cell-surface-receptor signalling. Nature Rev. Mol. Cell Biol. 3, 673–684 (2002).

    CAS  Google Scholar 

  5. Selkoe, D. & Kopan, R. Notch and presenilin: regulated intramembrane proteolysis links development and degeneration. Annu. Rev. Neurosci. 26, 565–597 (2003).

    CAS  PubMed  Google Scholar 

  6. Mumm, J. S. & Kopan, R. Notch signaling: from the outside in. Dev. Biol. 228, 151–165 (2000).

    CAS  PubMed  Google Scholar 

  7. Louvi, A. & Artavanis-Tsakonas, S. Notch signalling in vertebrate neural development. Nature Rev. Neurosci. 7, 93–102 (2006).

    CAS  Google Scholar 

  8. Lowell, S., Benchoua, A., Heavey, B. & Smith, A. G. Notch promotes neural lineage entry by pluripotent embryonic stem cells. PLoS Biol. 4, e121 (2006).

    PubMed  PubMed Central  Google Scholar 

  9. Fre, S. et al. Notch signals control the fate of immature progenitor cells in the intestine. Nature 435, 964–968 (2005).

    CAS  PubMed  Google Scholar 

  10. van Es, J. H. et al. Notch/γ-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 435, 959–963 (2005).

    CAS  PubMed  Google Scholar 

  11. Ohlstein, B. & Spradling, A. The adult Drosophila posterior midgut is maintained by pluripotent stem cells. Nature 439, 470–474 (2006).

    CAS  PubMed  Google Scholar 

  12. Micchelli, C. A. & Perrimon, N. Evidence that stem cells reside in the adult Drosophila midgut epithelium. Nature 439, 475–479 (2006).

    CAS  PubMed  Google Scholar 

  13. Weng, A. P. et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 306, 269–271 (2004). The analysis shows that 50% of patients with acute lymphoblastic leukaemia have activating mutations in the Notch1 locus. These map to two specific regions of the protein and confirm that the sequences close to the membrane in the ECD impose constraints that prevent premature activation.

    CAS  PubMed  Google Scholar 

  14. Chitnis, A. Why is Delta endocytosis required for effective activation of notch? Dev. Dyn. 235, 886–894 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Le Borgne, R., Bardin, A. & Schweisguth, F. The roles of receptor and ligand endocytosis in regulating Notch signaling. Development 132, 1751–1762 (2005).

    CAS  PubMed  Google Scholar 

  16. Itoh, M. et al. Mind bomb is a ubiquitin ligase that is essential for efficient activation of Notch signaling by Delta. Dev. Cell 4, 67–82 (2003). One of several papers reporting the importance of E3 ubiquitin ligases in regulating Notch ligands (see references 17–23). This is the first to identify Mind bomb. It also demonstrates the importance of ubiquitylation in promoting ligand activity in the signal-sending cell.

    CAS  PubMed  Google Scholar 

  17. Lai, E. C., Deblandre, G. A., Kintner, C. & Rubin, G. M. Drosophila neuralized is a ubiquitin ligase that promotes the internalization and degradation of Delta. Dev. Cell 1, 783–794 (2001).

    CAS  PubMed  Google Scholar 

  18. Pavlopoulos, E. et al. neuralized encodes a peripheral membrane protein involved in Delta signaling and endocytosis. Dev. Cell 1, 807–816 (2001).

    CAS  PubMed  Google Scholar 

  19. Deblandre, G. A., Lai, E. C. & Kintner, C. Xenopus neuralized is a ubiquitin ligase that interacts with XDelta1 and regulates Notch signaling. Dev. Cell 1, 795–806 (2001).

    CAS  PubMed  Google Scholar 

  20. Pitsouli, C. & Delidakis, C. The interplay between DSL proteins and ubiquitin ligases in Notch signaling. Development 132, 4041–4050 (2005).

    CAS  PubMed  Google Scholar 

  21. Le Borgne, R., Remaud, S., Hamel, S. & Schweisguth, F. Two distinct E3 ubiquitin ligases have complementary functions in the regulation of Delta and Serrate signaling in Drosophila. PLoS Biol. 3, e96 (2005).

    PubMed  PubMed Central  Google Scholar 

  22. Lai, E. C., Roegiers, F., Qin, X., Jan, Y. N. & Rubin, G. M. The ubiquitin ligase Drosophila Mind bomb promotes Notch signaling by regulating the localization and activity of Serrate and Delta. Development 132, 2319–2332 (2005).

    CAS  PubMed  Google Scholar 

  23. Wang, W. & Struhl, G. Distinct roles for Mind bomb, Neuralized and Epsin in mediating DSL endocytosis and signaling in Drosophila. Development 132, 2883–2894 (2005).

    CAS  PubMed  Google Scholar 

  24. Wang, W. & Struhl, G. Drosophila Epsin mediates a select endocytic pathway that DSL ligands must enter to activate Notch. Development 131, 5367–5380 (2004).

    CAS  PubMed  Google Scholar 

  25. Overstreet, E., Fitch, E. & Fischer, J. A. Fat facets and liquid facets promote Delta endocytosis and Delta signaling in the signaling cells. Development 131, 5355–5366 (2004).

    CAS  PubMed  Google Scholar 

  26. Hagedorn, E. J. et al. Drosophila melanogaster auxilin regulates the internalization of Delta to control activity of the Notch signaling pathway. J. Cell Biol. 173, 443–452 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Parks, A. L., Klueg, K. M., Stout, J. R. & Muskavitch, M. A. Ligand endocytosis drives receptor dissociation and activation in the Notch pathway. Development 127, 1373–1385 (2000).

    CAS  PubMed  Google Scholar 

  28. Hicks, C. et al. A secreted Delta1–Fc fusion protein functions both as an activator and inhibitor of Notch1 signaling. J. Neurosci. Res. 68, 655–667 (2002).

    CAS  PubMed  Google Scholar 

  29. Varnum-Finney, B. et al. Immobilization of Notch ligand, Delta-1, is required for induction of notch signaling. J. Cell Sci. 113, 4313–4318 (2000).

    CAS  PubMed  Google Scholar 

  30. Jafar-Nejad, H. et al. Sec15, a component of the exocyst, promotes notch signaling during the asymmetric division of Drosophila sensory organ precursors. Dev. Cell 9, 351–363 (2005).

    CAS  PubMed  Google Scholar 

  31. Emery, G. et al. Asymmetric Rab11 endosomes regulate Delta recycling and specify cell fate in the Drosophila nervous system. Cell 122, 763–773 (2005).

    CAS  PubMed  Google Scholar 

  32. Chen, N. & Greenwald, I. The lateral signal for LIN-12/Notch in C. elegans vulval development comprises redundant secreted and transmembrane DSL proteins. Dev. Cell 6, 183–192 (2004).

    CAS  PubMed  Google Scholar 

  33. Tian, X., Hansen, D., Schedl, T. & Skeath, J. B. Epsin potentiates Notch pathway activity in Drosophila and C. elegans. Development 131, 5807–5815 (2004).

    CAS  PubMed  Google Scholar 

  34. Bardin, A. J. & Schweisguth, F. Bearded family members inhibit neuralized-mediated endocytosis and signaling activity of Delta in Drosophila. Dev. Cell 10, 245–255 (2006).

    CAS  PubMed  Google Scholar 

  35. De Renzis, S., Yu, J., Zinzen, R. & Wieschaus, E. Dorsal-ventral pattern of Delta trafficking is established by a Snail–Tom–Neuralized pathway. Dev. Cell 10, 257–264 (2006). References 34 and 35 are the first to demonstrate that the Bearded-family members are inhibitors of the Neur E3 ligases. This has important implications for feedback regulation of Notch signalling.

    CAS  PubMed  Google Scholar 

  36. Escudero, L. M., Wei, S. Y., Chiu, W. H., Modolell, J. & Hsu, J. C. Echinoid synergizes with the Notch signaling pathway in Drosophila mesothorax bristle patterning. Development 130, 6305–6316 (2003).

    CAS  PubMed  Google Scholar 

  37. De Joussineau, C. et al. Delta-promoted filopodia mediate long-range lateral inhibition in Drosophila. Nature 426, 555–559 (2003).

    CAS  PubMed  Google Scholar 

  38. Wright, G. J., Leslie, J. D., Ariza-McNaughton, L. & Lewis, J. Delta proteins and MAGI proteins: an interaction of Notch ligands with intracellular scaffolding molecules and its significance for zebrafish development. Development 131, 5659–5669 (2004).

    CAS  PubMed  Google Scholar 

  39. Ascano, J. M., Beverly, L. J. & Capobianco, A. J. The C-terminal PDZ-ligand of JAGGED1 is essential for cellular transformation. J. Biol. Chem. 278, 8771–8779 (2003).

    CAS  PubMed  Google Scholar 

  40. Pfister, S. et al. Interaction of the MAGUK family member Acvrinp1 and the cytoplasmic domain of the Notch ligand Delta1. J. Mol. Biol. 333, 229–235 (2003).

    CAS  PubMed  Google Scholar 

  41. Klueg, K. M., Parody, T. R. & Muskavitch, M. A. Complex proteolytic processing acts on Delta, a transmembrane ligand for Notch, during Drosophila development. Mol. Biol. Cell 9, 1709–1723 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Qi, H. et al. Processing of the notch ligand Delta by the metalloprotease Kuzbanian. Science 283, 91–94 (1999).

    CAS  PubMed  Google Scholar 

  43. Mishra-Gorur, K., Rand, M. D., Perez-Villamil, B. & Artavanis-Tsakonas, S. Down-regulation of Delta by proteolytic processing. J. Cell Biol. 159, 313–324 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Sapir, A., Assa-Kunik, E., Tsruya, R., Schejter, E. & Shilo, B. Z. Unidirectional Notch signaling depends on continuous cleavage of Delta. Development 132, 123–132 (2005).

    CAS  PubMed  Google Scholar 

  45. Sun, X. & Artavanis-Tsakonas, S. Secreted forms of DELTA and SERRATE define antagonists of Notch signaling in Drosophila. Development 124, 3439–3448 (1997).

    CAS  PubMed  Google Scholar 

  46. Brooker, R., Hozumi, K. & Lewis, J. Notch ligands with contrasting functions: Jagged1 and Delta1 in the mouse inner ear. Development 133, 1277–1286 (2006).

    CAS  PubMed  Google Scholar 

  47. Irvine, K. D. Fringe, Notch, and making developmental boundaries. Curr. Opin. Genet. Dev. 9, 434–441 (1999).

    CAS  PubMed  Google Scholar 

  48. Haines, N. & Irvine, K. D. Glycosylation regulates Notch signalling. Nature Rev. Mol. Cell Biol. 4, 786–797 (2003).

    CAS  Google Scholar 

  49. Shi, S. & Stanley, P. Protein O-fucosyltransferase 1 is an essential component of Notch signaling pathways. Proc. Natl Acad. Sci. USA 100, 5234–5239 (2003).

    CAS  PubMed  Google Scholar 

  50. Sasamura, T. et al. neurotic, a novel maternal neurogenic gene, encodes an O-fucosyltransferase that is essential for Notch–Delta interactions. Development 130, 4785–4795 (2003).

    CAS  PubMed  Google Scholar 

  51. Okajima, T. & Irvine, K. D. Regulation of Notch signaling by O-linked fucose. Cell 111, 893–904 (2002).

    CAS  PubMed  Google Scholar 

  52. Okajima, T., Xu, A., Lei, L. & Irvine, K. D. Chaperone activity of protein O-fucosyltransferase 1 promotes notch receptor folding. Science 307, 1599–1603 (2005).

    CAS  PubMed  Google Scholar 

  53. Li, Y., Lei, L., Irvine, K. D. & Baker, N. E. Notch activity in neural cells triggered by a mutant allele with altered glycosylation. Development 130, 2829–2840 (2003).

    CAS  PubMed  Google Scholar 

  54. Bruckner, K., Perez, L., Clausen, H. & Cohen, S. Glycosyltransferase activity of Fringe modulates Notch–Delta interactions. Nature 406, 411–415 (2000).

    CAS  PubMed  Google Scholar 

  55. Moloney, D. J. et al. Fringe is a glycosyltransferase that modifies Notch. Nature 406, 369–375 (2000).

    CAS  PubMed  Google Scholar 

  56. Okajima, T., Xu, A. & Irvine, K. D. Modulation of Notch-ligand binding by protein O-fucosyltransferase 1 and fringe. J. Biol. Chem. 278, 42340–42345 (2003).

    CAS  PubMed  Google Scholar 

  57. Lei, L., Xu, A., Panin, V. M. & Irvine, K. D. An O-fucose site in the ligand binding domain inhibits Notch activation. Development 130, 6411–6421 (2003).

    CAS  PubMed  Google Scholar 

  58. Yang, L. T. et al. Fringe glycosyltransferases differentially modulate Notch1 proteolysis induced by Delta1 and Jagged1. Mol. Biol. Cell 16, 927–942 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Pourquie, O. The segmentation clock: converting embryonic time into spatial pattern. Science 301, 328–330 (2003).

    CAS  PubMed  Google Scholar 

  60. Sato, Y., Yasuda, K. & Takahashi, Y. Morphological boundary forms by a novel inductive event mediated by Lunatic fringe and Notch during somitic segmentation. Development 129, 3633–3644 (2002).

    CAS  PubMed  Google Scholar 

  61. Struhl, G. & Adachi, A. Requirements for presenilin-dependent cleavage of Notch and other transmembrane proteins. Mol. Cell 6, 625–636 (2000).

    CAS  PubMed  Google Scholar 

  62. Fortini, M. E. Notch and presenilin: a proteolytic mechanism emerges. Curr. Opin. Cell Biol. 13, 627–634 (2001).

    CAS  PubMed  Google Scholar 

  63. Mumm, J. S. et al. A ligand-induced extracellular cleavage regulates γ-secretase-like proteolytic activation of Notch1. Mol. Cell 5, 197–206 (2000). Using a combination of biochemical and pharmacological approaches, combined with Notch constructs that mimic different activating mutations, this paper is the first to posit that Notch activation involves a proteolytic cascade. The accompanying paper (reference 64) also identified the S2 cleavage.

    CAS  PubMed  Google Scholar 

  64. Brou, C. et al. A novel proteolytic cleavage involved in Notch signaling: the role of the disintegrin-metalloprotease TACE. Mol. Cell 5, 207–216 (2000).

    CAS  PubMed  Google Scholar 

  65. Jarriault, S. & Greenwald, I. Evidence for functional redundancy between C. elegans ADAM proteins SUP-17/Kuzbanian and ADM-4/TACE. Dev. Biol. 287, 1–10 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Huovila, A. P., Turner, A. J., Pelto-Huikko, M., Karkkainen, I. & Ortiz, R. M. Shedding light on ADAM metalloproteinases. Trends Biochem. Sci. 30, 413–422 (2005).

    CAS  PubMed  Google Scholar 

  67. Gupta-Rossi, N. et al. Monoubiquitination and endocytosis direct γ-secretase cleavage of activated Notch receptor. J. Cell Biol. 166, 73–83 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Herranz, H., Stamataki, E., Feiguin, F. & Milan, M. Self-refinement of Notch activity through the transmembrane protein Crumbs: modulation of γ-Secretase activity. EMBO Rep. 7, 297–302 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Aster, J. C. Deregulated NOTCH signaling in acute T-cell lymphoblastic leukemia/lymphoma: new insights, questions, and opportunities. Int. J. Hematol. 82, 295–301 (2005).

    CAS  PubMed  Google Scholar 

  70. Wilkin, M. B. et al. Regulation of notch endosomal sorting and signaling by Drosophila Nedd4 family proteins. Curr. Biol. 14, 2237–2244 (2004).

    CAS  PubMed  Google Scholar 

  71. Jekely, G. & Rorth, P. Hrs mediates downregulation of multiple signalling receptors in Drosophila. EMBO Rep. 4, 1163–1168 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Lu, H. & Bilder, D. Endocytic control of epithelial polarity and proliferation in Drosophila. Nature Cell Biol. 7, 1132–1139 (2005).

    CAS  Google Scholar 

  73. Mukherjee, A. et al. Regulation of Notch signalling by non-visual β-arrestin. Nature Cell Biol. 7, 1191–1201 (2005).

    PubMed  Google Scholar 

  74. Thompson, B. J. et al. Tumor suppressor properties of the ESCRT-II complex component Vps25 in Drosophila. Dev. Cell 9, 711–720 (2005).

    CAS  PubMed  Google Scholar 

  75. Vaccari, T. & Bilder, D. The Drosophila tumor suppressor vps25 prevents nonautonomous overproliferation by regulating notch trafficking. Dev. Cell 9, 687–698 (2005).

    CAS  PubMed  Google Scholar 

  76. Moberg, K. H., Schelble, S., Burdick, S. K. & Hariharan, I. K. Mutations in erupted, the Drosophila ortholog of mammalian tumor susceptibility gene 101, elicit non-cell-autonomous overgrowth. Dev. Cell 9, 699–710 (2005).

    CAS  PubMed  Google Scholar 

  77. Berdnik, D., Torok, T., Gonzalez-Gaitan, M. & Knoblich, J. A. The endocytic protein α-adaptin is required for numb-mediated asymmetric cell division in Drosophila. Dev. Cell 3, 221–231 (2002). By showing that Numb interacts with α-adaptin this paper makes an important link between Numb and endocytosis.

    CAS  PubMed  Google Scholar 

  78. McGill, M. A. & McGlade, C. J. Mammalian numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain. J. Biol. Chem. 278, 23196–23203 (2003).

    CAS  PubMed  Google Scholar 

  79. O'Connor-Giles, K. M. & Skeath, J. B. Numb inhibits membrane localization of Sanpodo, a four-pass transmembrane protein, to promote asymmetric divisions in Drosophila. Dev. Cell 5, 231–243 (2003).

    CAS  PubMed  Google Scholar 

  80. Hutterer, A. & Knoblich, J. A. Numb and α-adaptin regulate Sanpodo endocytosis to specify cell fate in Drosophila external sensory organs. EMBO Rep. 6, 836–842 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Chien, C. T., Wang, S., Rothenberg, M., Jan, L. Y. & Jan, Y. N. Numb-associated kinase interacts with the phosphotyrosine binding domain of Numb and antagonizes the function of Numb in vivo. Mol. Cell. Biol. 18, 598–607 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Fares, H. & Greenwald, I. SEL-5, a serine/threonine kinase that facilitates LIN-12 activity in Caenorhabditis elegans. Genetics 153, 1641–1654 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Tang, H. et al. Numb proteins specify asymmetric cell fates via an endocytosis- and proteasome-independent pathway. Mol. Cell. Biol. 25, 2899–2909 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Lai, E. C. Protein degradation: four E3s for the notch pathway. Curr. Biol. 12, R74–R78 (2002).

    CAS  PubMed  Google Scholar 

  85. Qiu, L. et al. Recognition and ubiquitination of Notch by Itch, a hect-type E3 ubiquitin ligase. J. Biol. Chem. 275, 35734–35737 (2000).

    CAS  PubMed  Google Scholar 

  86. Shaye, D. D. & Greenwald, I. Endocytosis-mediated downregulation of LIN-12/Notch upon Ras activation in Caenorhabditis elegans. Nature 420, 686–690 (2002). This is one of the clearest examples in which regulation of Notch endocytosis is important in the appropriate specification of cell fates in vivo.

    CAS  PubMed  Google Scholar 

  87. Shaye, D. D. & Greenwald, I. LIN-12/Notch trafficking and regulation of DSL ligand activity during vulval induction in Caenorhabditis elegans. Development 132, 5081–5092 (2005).

    CAS  PubMed  Google Scholar 

  88. Sakata, T. et al. Drosophila Nedd4 regulates endocytosis of Notch and suppresses its ligand-independent activation. Curr. Biol. 14, 2228–2236 (2004).

    CAS  PubMed  Google Scholar 

  89. Gallagher, E., Gao, M., Liu, Y. C. & Karin, M. Activation of the E3 ubiquitin ligase Itch through a phosphorylation-induced conformational change. Proc. Natl Acad. Sci. USA 103, 1717–1722 (2006).

    CAS  PubMed  Google Scholar 

  90. Matsuno, K., Diederich, R. J., Go, M. J., Blaumueller, C. M. & Artavanis-Tsakonas, S. Deltex acts as a positive regulator of Notch signaling through interactions with the Notch ankyrin repeats. Development 121, 2633–2644 (1995).

    CAS  PubMed  Google Scholar 

  91. Hori, K. et al. Drosophila Deltex mediates Suppressor of Hairless-independent and late-endosomal activation of Notch signaling. Development 131, 5527–5537 (2004).

    CAS  PubMed  Google Scholar 

  92. Sestan, N., Artavanis-Tsakonas, S. & Rakic, P. Contact-dependent inhibition of cortical neurite growth mediated by Notch signaling. Science 286, 741–746 (1999).

    CAS  PubMed  Google Scholar 

  93. Kovall, R. A. & Hendrickson, W. A. Crystal structure of the nuclear effector of Notch signaling, CSL, bound to DNA. EMBO J. 23, 3441–3451 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Nam, Y., Sliz, P., Song, L., Aster, J. C. & Blacklow, S. C. Structural basis for cooperativity in recruitment of MAML coactivators to Notch transcription complexes. Cell 124, 973–983 (2006).

    CAS  PubMed  Google Scholar 

  95. Wilson, J. J. & Kovall, R. A. Crystal structure of the CSL–Notch–Mastermind ternary complex bound to DNA. Cell 124, 985–996 (2006). The crystal structure described in references 94 and 95 show for the first time the interactions between the key proteins in the Nicd-transcription-activation complex.

    CAS  PubMed  Google Scholar 

  96. Petcherski, A. G. & Kimble, J. Mastermind is a putative activator for Notch. Curr. Biol. 10, R471–R473 (2000).

    CAS  PubMed  Google Scholar 

  97. Wu, L. et al. MAML1, a human homologue of Drosophila mastermind, is a transcriptional co-activator for NOTCH receptors. Nature Genet. 26, 484–489 (2000).

    CAS  PubMed  Google Scholar 

  98. Fryer, C. J., White, J. B. & Jones, K. A. Mastermind recruits CycC–CDK8 to phosphorylate the Notch ICD and coordinate activation with turnover. Mol. Cell 16, 509–520 (2004). This paper makes an important link between Nicd function on the DNA and its turnover. Their results show co-recruitment of a kinase to Notch-target enhancer where it phosphorylates Nicd and promotes ubiquitin-mediated turnover.

    CAS  PubMed  Google Scholar 

  99. Zhou, S. et al. SKIP, a CBF1-associated protein, interacts with the ankyrin repeat domain of NotchIC to facilitate NotchIC function. Mol. Cell. Biol. 20, 2400–2410 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Wallberg, A. E., Pedersen, K., Lendahl, U. & Roeder, R. G. p300 and PCAF act cooperatively to mediate transcriptional activation from chromatin templates by notch intracellular domains in vitro. Mol. Cell. Biol. 22, 7812–7819 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Fryer, C. J., Lamar, E., Turbachova, I., Kintner, C. & Jones, K. A. Mastermind mediates chromatin-specific transcription and turnover of the Notch enhancer complex. Genes Dev. 16, 1397–1411 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Gupta-Rossi, N. et al. Functional interaction between SEL-10, an F-box protein, and the nuclear form of activated Notch1 receptor. J. Biol. Chem. 276, 34371–34378 (2001).

    CAS  PubMed  Google Scholar 

  103. Wu, G. et al. SEL-10 is an inhibitor of notch signaling that targets notch for ubiquitin-mediated protein degradation. Mol. Cell. Biol. 21, 7403–7015 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Oberg, C. et al. The Notch intracellular domain is ubiquitinated and negatively regulated by the mammalian Sel-10 homolog. J. Biol. Chem. 276, 35847–35853 (2001).

    CAS  PubMed  Google Scholar 

  105. Nagel, A. C. et al. Hairless-mediated repression of Notch target genes requires the combined activity of Groucho and CtBP corepressors. Mol. Cell. Biol. 25, 10433–10441 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Morel, V. et al. Transcriptional repression by Suppressor of Hairless involves the binding of a hairless–dCtBP complex in Drosophila. Curr. Biol. 11, 789–792 (2001).

    CAS  PubMed  Google Scholar 

  107. Castro, B., Barolo, S., Bailey, A. M. & Posakony, J. W. Lateral inhibition in proneural clusters: cis-regulatory logic and default repression by Suppressor of Hairless. Development 132, 3333–3344 (2005).

    CAS  PubMed  Google Scholar 

  108. Kao, H. Y. et al. A histone deacetylase corepressor complex regulates the Notch signal transduction pathway. Genes Dev. 12, 2269–2277 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Oswald, F. et al. RBP-Jκ/SHARP recruits CtIP/CtBP corepressors to silence Notch target genes. Mol. Cell. Biol. 25, 10379–10390 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Hsieh, J. J., Zhou, S., Chen, L., Young, D. B. & Hayward, S. D. CIR, a corepressor linking the DNA binding factor CBF1 to the histone deacetylase complex. Proc. Natl Acad. Sci. USA 96, 23–28 (1999).

    CAS  PubMed  Google Scholar 

  111. Tsuda, L., Nagaraj, R., Zipursky, S. L. & Banerjee, U. An EGFR–Ebi–Sno pathway promotes Delta expression by inactivating Su(H)/SMRTER repression during inductive notch signaling. Cell 110, 625–637 (2002).

    CAS  PubMed  Google Scholar 

  112. Kuang, B., Wu, S. C., Shin, Y., Luo, L. & Kolodziej, P. split ends encodes large nuclear proteins that regulate neuronal cell fate and axon extension in the Drosophila embryo. Development 127, 1517–1529 (2000).

    CAS  PubMed  Google Scholar 

  113. Morel, V. & Schweisguth, F. Repression by Suppressor of Hairless and activation by Notch are required to define a single row of Single-minded expressing cells in the Drosophila embryo. Genes Dev. 14, 377–388 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Barolo, S., Stone, T., Bang, A. G. & Posakony, J. W. Default repression and Notch signaling: Hairless acts as an adaptor to recruit the corepressors Groucho and dCtBP to Suppressor of Hairless. Genes Dev 16, 1964–1976 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Bray, S. & Furriols, M. Notch pathway: making sense of Suppressor of hairless. Curr. Biol. 11, R217–R221 (2001).

    CAS  PubMed  Google Scholar 

  116. Koelzer, S. & Klein, T. Regulation of expression of Vg and establishment of the dorsoventral compartment boundary in the wing imaginal disc by Suppressor of Hairless. Dev. Biol. 289, 77–90 (2006).

    CAS  PubMed  Google Scholar 

  117. Koelzer, S. & Klein, T. A Notch-independent function of Suppressor of Hairless during the development of the bristle sensory organ precursor cell of Drosophila. Development 130, 1973–1988 (2003).

    CAS  PubMed  Google Scholar 

  118. Barolo, S. et al. A Notch-independent activity of Suppressor of hairless is required for normal mechanoreceptor physiology. Cell 103, 957–969 (2000).

    CAS  PubMed  Google Scholar 

  119. Kadam, S. & Emerson, B. M. Transcriptional specificity of human SWI/SNF BRG1 and BRM chromatin remodeling complexes. Mol. Cell 11, 377–389 (2003).

    CAS  PubMed  Google Scholar 

  120. Kurooka, H. & Honjo, T. Functional interaction between the mouse Notch1 intracellular region and histone acetyltransferases PCAF and GCN5. J. Biol. Chem. 275, 17211–17220 (2000).

    CAS  PubMed  Google Scholar 

  121. Armstrong, J. A. et al. Genetic screens for enhancers of Brahma reveal functional interactions between the BRM chromatin-remodeling complex and the Delta–Notch signal transduction pathway in Drosophila. Genetics 170, 1761–1774 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Gause, M. et al. Nipped-A, the Tra1/TRRAP subunit of the Drosophila SAGA and Tip60 complexes, has multiple roles in Notch signaling during wing development. Mol. Cell. Biol. 26, 2347–2359 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Bray, S., Musisi, H. & Bienz, M. Bre1 is required for Notch signaling and histone modification. Dev. Cell 8, 279–286 (2005).

    CAS  PubMed  Google Scholar 

  124. Poulin, G., Dong, Y., Fraser, A. G., Hopper, N. A. & Ahringer, J. Chromatin regulation and sumoylation in the inhibition of Ras-induced vulval development in Caenorhabditis elegans. EMBO J. 24, 2613–2623 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Ferres-Marco, D. et al. Epigenetic silencers and Notch collaborate to promote malignant tumours by Rb silencing. Nature 439, 430–436 (2006).

    CAS  PubMed  Google Scholar 

  126. Furriols, M. & Bray, S. A model Notch response element detects Suppressor of Hairless-dependent molecular switch. Curr. Biol. 11, 60–64 (2001).

    CAS  PubMed  Google Scholar 

  127. Cooper, M. T. et al. Spatially restricted factors cooperate with notch in the regulation of Enhancer of Split genes. Dev. Biol. 221, 390–403 (2000).

    CAS  PubMed  Google Scholar 

  128. Cave, J. W., Loh, F., Surpris, J. W., Xia, L. & Caudy, M. A. A DNA transcription code for cell-specific gene activation by notch signaling. Curr. Biol. 15, 94–104 (2005).

    CAS  PubMed  Google Scholar 

  129. Ong, C. T. et al. Target selectivity of vertebrate notch proteins. Collaboration between discrete domains and CSL-binding site architecture determines activation probability. J. Biol. Chem. 281, 5106–5119 (2006).

    CAS  PubMed  Google Scholar 

  130. Giudicelli, F. & Lewis, J. The vertebrate segmentation clock. Curr. Opin. Genet. Dev. 14, 407–414 (2004).

    CAS  PubMed  Google Scholar 

  131. Langevin, J. et al. Lethal giant larvae controls the localization of Notch-signaling regulators Numb, Neuralized, and Sanpodo in Drosophila sensory-organ precursor cells. Curr. Biol. 15, 955–962 (2005).

    CAS  PubMed  Google Scholar 

  132. Hirata, H. et al. Instability of Hes7 protein is crucial for the somite segmentation clock. Nature Genet. 36, 750–754 (2004).

    CAS  PubMed  Google Scholar 

  133. Stark, A., Brennecke, J., Russell, R. B. & Cohen, S. M. Identification of Drosophila MicroRNA targets. PLoS Biol. 1, E60 (2003).

    PubMed  PubMed Central  Google Scholar 

  134. Lai, E. C., Tam, B. & Rubin, G. M. Pervasive regulation of Drosophila Notch target genes by GY-box-, Brd-box-, and K-box-class microRNAs. Genes Dev. 19, 1067–1080 (2005). This paper demonstrates that three different families of Drosophila miRNAs directly regulate two large families of Notch target genes.

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Neves, A. & Priess, J. R. The REF-1 family of bHLH transcription factors pattern C. elegans embryos through Notch-dependent and Notch-independent pathways. Dev. Cell 8, 867–879 (2005).

    CAS  PubMed  Google Scholar 

  136. Le Borgne, R. & Schweisguth, F. Unequal segregation of Neuralized biases Notch activation during asymmetric cell division. Dev. Cell 5, 139–148 (2003).

    CAS  PubMed  Google Scholar 

  137. Cheng, Y. C. et al. Notch activation regulates the segregation and differentiation of rhombomere boundary cells in the zebrafish hindbrain. Dev. Cell 6, 539–550 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

My apologies to all those colleagues whose important contributions have not been acknowledged due to the space constraints of this Review. My thanks to A. Krejci, M. Glittenberg, A. Djiane and the reviewers for helpful comments, and to F. Wirtz-Peitz and J. Knoblich (Institute of Molecular Biotechnology, Vienna, Austria) for the beautiful image of SOPs in Box 2. Work on Notch signalling in my laboratory is currently supported by grants from the Medical Research Council, the Wellcome Trust and the Association for International Cancer Research.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

Related links

DATABASES

OMIN

Alzheimer's disease

FURTHER INFORMATION

Sarah Bray's homepage

Glossary

ADAM-family metalloproteases

Transmembrane disintegrins and metalloproteases that proteolytically cleave the juxtamembrane region of cellular transmembrane proteins and detach their extracellular regions — this process is known as ectodomain shedding.

γ-secretase complex

Presenilin, a multispan membrane protein, is the catalytic subunit, and the transmembrane proteins nicastrin and APH1 stabilize the presenilin holoprotein. PEN2, a two-pass transmembrane protein, induces endoproteolysis of presenilin and maturation of the γ-secretase complex.

E3 ubiquitin ligase

An adaptor protein that links ubiquitin-conjugating E2 enzymes with substrates and contributes to the catalytic transfer of ubiquitin onto the substrate.

Epsin

A clathrin and phosphatidylinositol-4,5-bisphosphate-binding protein that contains ubiquitin-interaction motifs. It is thought to facilitate endocytosis of ubiquitylated cargo proteins.

Auxilin

A J-domain-containing protein that is implicated in the disassembly of clathrin from clathrin-coated vesicles.

Recycling endosome

A compartment that sorts transmembrane proteins that are recycled to the plasma membrane following endocytosis.

Exocyst

A heteromeric protein complex that is required for polarized exocytosis of post-Golgi secretory vesicles.

Bearded-related proteins

Short polypeptides that were identified in insects and contain an N-terminal amphipathic helix and 2 or 3 conserved motifs.

Adherens junction

A cell–cell junction that mediates adhesion through cadherins and regulates and/or links to the actin cytoskeleton.

PDZ-binding motif

A motif at the C terminus of a protein that is recognized by a PDZ-domain-containing protein. PDZ-domains are conserved 80–90-residue domains that fold into a β-sandwich and are found in many scaffold and signalling proteins.

Fringe

A Golgi-resident glycosyl-transferase that was first identified in D. melanogaster and has three homologues in mammals: Lunatic Fringe, Radical Fringe and Manic Fringe.

RAB GTPases

Members of the Ras superfamily of small GTPases, RAB proteins regulate vesicle budding, fusion and motility.

HRS/VPS27

A protein that contains ubiquitin-interaction motifs and is important for sorting ubiquitylated endosomal cargoes.

Syntaxin I

Integral membrane protein with sequence similarity to t-SNAREs that is involved in vesicle docking and vesicle fusion.

β-arrestin

Also known as non-visual arrestin, it is a cytoplasmic protein that promotes endocytosis of G-protein-coupled receptors and is present in coated vesicles.

ESCRT

(Endosomal sorting complex required for transport). Three heteromeric protein complexes, ESCRTI, ESCRTII and ESCRTIII, function sequentially in the sorting of membrane proteins into the multivesicular body.

AP2 complex

A heterotetrameric trafficking adaptor complex that comprises two large, one medium and one small subunit. It interacts with clathrin and also, through the appendage domain of a large subunit, binds to accessory proteins, including Epsins.

Rel family

Also known as nuclear factor (NF)-κB proteins, this family of transcription factors contains a conserved domain (the Rel-homology domain (RHD)) that is required for DNA binding and dimerization. These proteins are important in defence against infectious diseases and cellular stress.

Somitogenesis

The development of somites, the segmental blocks of mesoderm that give rise to the axial skeleton, muscles and dermis.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bray, S. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol 7, 678–689 (2006). https://doi.org/10.1038/nrm2009

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm2009

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