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:

Signal transduction and endocytosis: close encounters of many kinds

Key Points

  • Many receptor tyrosine kinases (RTKs) and G-protein-coupled receptors (GPCRs) are rapidly endocytosed after ligand-induced activation, and then move through a series of endosomal compartments. Receptors can be recycled to the plasma membrane after endocytosis or can be retained in multivesicular bodies (MVBs), trapped in the MVB interior and delivered to lysosomes.

  • Endocytosis has an essential role in controlling the amounts of signalling receptors and their ligands on the cell surface and extracellular medium, thereby reducing or elevating the intensity of receptor signalling. During development, the endocytic machinery acts in various ways to regulate signalling by soluble factors and membrane factors by establishing gradients and regulating growth-factor accessibility.

  • Many growth factors remain bound to endocytosed receptors, which maintains the activity of the receptors in endosomes. This sustained activation allows signalling proteins to bind receptors in endosomes. Several downstream signalling proteins that are involved in MAPK pathways are also found in endosomes that contain internalized RTKs and GPCRs.

  • In neuronal cells, endocytosed TrkA receptors remain activated in endosomes. TrkA receptors that are activated and internalized in the axonal terminals signal to MAPKs not only locally in axons but also in the cell body in which they can be transported in endosomes.

  • Inhibition of endocytosis of RTKs or GPCRs by dominant-negative mutants of endocytic proteins causes impaired activation of MAPK pathways in some experimental systems, which indicates that endocytosis is essential for MAPK activation.

  • Activation of signalling receptors leads to phosphorylation and ubiquitylation of several components of the clathrin coat. These modifications might contribute to the specific mechanisms of ligand-induced endocytosis of RTKs and GPCRs.

  • Signal-induced ubiquitylation of endosomal proteins and activation of endosomal enzymes have an essential role in the sorting of RTKs and GPCRs to the lysosomal degradation pathway. In MVBs, ubiquitylated receptors interact with proteins carrying ubiquitin-interacting motifs, which leads to trapping the receptors into internal vesicles of MVBs.

Abstract

Binding of hormones, growth factors and other cell modulators to cell-surface receptors triggers a complex array of signal-transduction events. The activation of many receptors also accelerates their endocytosis. Endocytic transport is important in regulating signal transduction and in mediating the formation of specialized signalling complexes. Conversely, signal-transduction events modulate specific components of the endocytic machinery. Recent studies of protein tyrosine kinases and G-protein-coupled receptors have shed new light on the mechanisms and functional consequences of this bidirectional interplay between signalling and membrane-transport networks.

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: Endocytic transport itineraries of receptor tyrosine kinases.
Figure 2: Endocytic transport itineraries of G-protein-coupled receptors.
Figure 3: Desensitization and resensitization of G-protein-coupled receptors during endocytosis.
Figure 4: Role of endocytosis in Notch signalling in Drosophila.
Figure 5: Termination of receptor-tyrosine-kinase signalling in multivesicular bodies.
Figure 6: Hypothetical model of signalling complexes in endosomes.
Figure 7: Retrograde transport of endosomes containing TrkA signalling complexes in neurons.
Figure 8: Endocytic protein complexes modified by signalling receptors.
Figure 9: Protein complexes modified by signalling receptors in endosomes.

Similar content being viewed by others

References

  1. Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 103, 211–225 (2000).

    CAS  PubMed  Google Scholar 

  2. Neer, E. J. Heterotrimeric G proteins: organizers of transmembrane signals. Cell 80, 249–257 (1995).

    CAS  PubMed  Google Scholar 

  3. Gorden, P., Carpentier, J.-L., Cohen, S. & Orci, L. Epidermal growth factor: morphological demonstration of binding internalization and lysosomal association in human fibroblasts. Proc. Natl Acad. Sci. USA 75, 5025–5029 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Hanover, J. A., Willingham, M. C. & Pastan, I. Kinetics of transit of transferrin and epidermal growth factor through clathrin-coated membranes. Cell 39, 283–293 (1984).

    CAS  PubMed  Google Scholar 

  5. Carpentier, J.-L. et al. Co-localization of 125I-epidermal growth factor and ferritin-low density lipoprotein in coated pits: a quantitative electron microscopic study in normal and mutant human fibroblasts. J. Cell Biol. 95, 73–77 (1982).

    CAS  PubMed  Google Scholar 

  6. Damke, H., Baba, T., Warnock, D. E. & Schmid, S. L. Induction of mutant dynamin specifically blocks endocytic coated vesicle formation. J. Cell Biol. 127, 915–934 (1994).

    CAS  PubMed  Google Scholar 

  7. Carbone, R. et al. eps15 and eps15R are essential components of the endocytic pathway. Cancer Res. 57, 5498–5504 (1997).

    CAS  PubMed  Google Scholar 

  8. Haigler, H. T., McKanna, J. A. & Cohen, S. Direct visualization of the binding and internalization of a ferritin conjugate of epidermal growth factor in human carcinoma cells A431. J. Cell Biol. 81, 382–395 (1979).

    CAS  PubMed  Google Scholar 

  9. Hopkins, C. R., Miller, K. & Beardmore, J. M. Receptor-mediated endocytosis of transferrin and epidermal growth factor receptors: a comparison of constitutive and ligand-induced uptake. J. Cell Sci. 3, 173–186 (1985).

    CAS  Google Scholar 

  10. Von Zastrow, M. & Kobilka, B. K. Ligand-regulated internalization and recycling of human β 2-adrenergic receptors between the plasma membrane and endosomes containing transferrin receptors. J. Biol. Chem. 267, 3530–3538 (1992).

    CAS  PubMed  Google Scholar 

  11. Goodman, O. B. Jr et al. β-arrestin acts as a clathrin adaptor in endocytosis of the β2- adrenergic receptor. Nature 383, 447–450 (1996).This study provided early evidence that β-arrestins, in addition to preventing GPCR interaction with heterotrimeric G proteins, can directly link GPCRs to clathrin-coated pits.

    CAS  PubMed  Google Scholar 

  12. Roettger, B. F. et al. Dual pathways of internalization of the cholecystokinin receptor. J. Cell Biol. 128, 1029–1041 (1995).

    CAS  PubMed  Google Scholar 

  13. Zhang, J., Ferguson, S., Barak, L. S., Menard, L. & Caron, M. G. Dynamin and β-arrestin reveal distinct mechanisms for G protein-coupled receptor internalization. J. Biol. Chem. 271, 18302–18305 (1996).

    CAS  PubMed  Google Scholar 

  14. Vickery, R. G. & von Zastrow, M. Distinct dynamin-dependent and -independent mechanisms target structurally homologous dopamine receptors to different endocytic membranes. J. Cell Biol. 144, 31–43 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Liggett, S. B., Freedman, N. J., Schwinn, D. A. & Lefkowitz, R. J. Structural basis for receptor subtype-specific regulation revealed by a chimeric β3/β2-adrenergic receptor. Proc. Natl Acad. Sci. USA 90, 3665–3669 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Von Zastrow, M., Link, R., Daunt, D., Barsh, G. & Kobilka, B. Subtype-specific differences in the intracellular sorting of G protein-coupled receptors. J. Biol. Chem. 268, 763–766 (1993).

    CAS  PubMed  Google Scholar 

  17. Roettger, B. F. et al. Insulation of a G protein-coupled receptor on the plasmalemmal surface of the pancreatic acinar cell. J. Cell Biol. 130, 579–590 (1995).

    CAS  PubMed  Google Scholar 

  18. Gruenberg, J. & Maxfield, F. R. Membrane transport in the endocytic pathway. Curr. Opin. Cell Biol. 7, 552–563 (1995).

    CAS  PubMed  Google Scholar 

  19. McKanna, J. A., Haigler, H. T. & Cohen, S. Hormone receptor topology and dynamics: morphological analysis using ferritin-labeled epidermal growth factor. Proc. Natl Acad. Sci. USA 76, 5689–5693 (1979).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Miller, K., Beardmore, J., Kanety, H., Schlessinger, J. & Hopkins, C. R. Localization of epidermal growth factor (EGF) receptor within the endosome of EGF-stimulated epidermoid carcinoma (A431) cells. J. Cell Biol. 102, 500–509 (1986).

    CAS  PubMed  Google Scholar 

  21. Sorkin, A. et al. Recycling of epidermal growth factor-receptor complexes in A431 cells: identification of dual pathways. J. Cell Biol. 112, 55–63 (1991).

    CAS  PubMed  Google Scholar 

  22. Felder, S. et al. Kinase activity controls the sorting epidermal growth factor receptor within the multivesicular body. Cell 61, 623–634 (1990).

    CAS  PubMed  Google Scholar 

  23. Ghinea, N. et al. Pathways of internalization of the hCG/LH receptor: immunoelectron microscopic studies in Leydig cells and transfected L-cells. J. Cell Biol. 118, 1347–1358 (1992).

    CAS  PubMed  Google Scholar 

  24. Volpicelli, L. A., Lah, J. J. & Levey, A. I. Rab5-dependent trafficking of the m4 muscarinic acetylcholine receptor to the plasma membrane, early endosomes, and multivesicular bodies. J. Biol. Chem. 276, 47590–47598 (2001).

    CAS  PubMed  Google Scholar 

  25. Stoscheck, C. M. & Carpenter, G. 'Down-regulation' of EGF receptors: direct demonstration of receptor degradation in human fibroblasts. J. Cell Biol. 98, 1048–1053 (1984).

    CAS  PubMed  Google Scholar 

  26. Beguinot, L., Lyall, R. M., Willingham, M. C. & Pastan, I. Down-regulation of the epidermal growth factor receptor in KB cells is due to receptor internalization and subsequent degradation in lysosomes. Proc. Natl Acad. Sci. USA 81, 2384–2388 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Wells, A. et al. Ligand-induced transformation by a noninternalizing epidermal growth factor receptor. Science 247, 962–964 (1990).The first demonstration of the role of RTK endocytosis in attenuation of the mitogenic signalling by growth factors.

    CAS  PubMed  Google Scholar 

  28. Tsao, P. & von Zastrow, M. Downregulation of G protein-coupled receptors. Curr. Opin. Neurobiol. 10, 365–369 (2000).

    CAS  PubMed  Google Scholar 

  29. Lefkowitz, R. J., Pitcher, J., Krueger, K. & Daaka, Y. Mechanisms of β-adrenergic receptor desensitization and resensitization. Adv. Pharmacol. 42, 416–420 (1998).

    CAS  PubMed  Google Scholar 

  30. Zhang, J. et al. Molecular mechanisms of G protein-coupled receptor signaling: role of G protein-coupled receptor kinases and arrestins in receptor desensitization and resensitization. Receptors Channels 5, 193–199 (1997).

    CAS  PubMed  Google Scholar 

  31. Dubois, L., Lecourtois, M., Alexandre, C., Hirst, E. & Vincent, J. P. Regulated endocytic routing modulates wingless signaling in Drosophila embryos. Cell 105, 613–624 (2001).

    CAS  PubMed  Google Scholar 

  32. Strigini, M. & Cohen, S. M. Wingless gradient formation in the Drosophila wing. Curr. Biol. 10, 293–300 (2000).

    CAS  PubMed  Google Scholar 

  33. Entchev, E. V., Schwabedissen, A. & Gonzalez-Gaitan, M. Gradient formation of the TGF-β homolog Dpp. Cell 103, 981–991 (2000).

    CAS  PubMed  Google Scholar 

  34. Srinivasan, S., Rashka, K. E. & Bier, E. Creation of a Sog morphogen gradient in the Drosophila embryo. Dev. Cell 2, 91–101 (2002).

    CAS  PubMed  Google Scholar 

  35. 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).This paper provided the first evidence for the role of endocytosis in regulation of Notch signalling during Drosophila development.

    CAS  PubMed  Google Scholar 

  36. 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 

  37. 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 

  38. Goldstein, J. L., Brown, M. S., Anderson, R. G. W., Russel, D. W. & Schneider, W. J. Receptor-mediated endocytosis: concepts emerging from the LDL receptor system. Annu. Rev. Cell Biol. 1, 1–19 (1985).

    CAS  PubMed  Google Scholar 

  39. Sorkin, A., Teslenko, L. & Nikolsky, N. The endocytosis of epidermal growth factor in A431 cells: a pH of microenvironment and the dynamics of receptor complexes dissociation. Exp. Cell Res. 175, 192–205 (1988).

    CAS  PubMed  Google Scholar 

  40. Sorkin, A., Eriksson, A., Heldin, C.-H., Westermark, B. & Claesson-Welsh, L. Pool of ligand-bound platelet-derived growth factor β-receptors remain activated and tyrosine-phosphorylated after internalization. J. Cell Physiol. 156, 373–382 (1993).

    CAS  PubMed  Google Scholar 

  41. Zapf-Colby, A. & Olefsky, J. M. Nerve growth factor processing and trafficking events following TrkA-mediated endocytosis. Endocrinology 139, 3232–3240 (1998).

    CAS  PubMed  Google Scholar 

  42. French, A. R., Tadaki, D. K., Niyogi, S. K. & Lauffenberger, D. A. Intracellular trafficking of epidermal growth factor family ligands is directly influenced by the pH sensitivity of the receptor/ligand interaction. J. Biol. Chem. 270, 4334–4340 (1995).

    CAS  PubMed  Google Scholar 

  43. Authier, F., Rachubinski, R. A., Posner, B. I. & Bergeron, J. J. Endosomal proteolysis of insulin by an acidic thiol metalloprotease unrelated to insulin degrading enzyme. J. Biol. Chem. 269, 3010–3016 (1994).

    CAS  PubMed  Google Scholar 

  44. Korc, M. & Magun, B. E. Binding and processing of epidermal growth factor in Panc-I human pancreatic carcinoma cells. Life Sci. 36, 1849–1855 (1985).

    CAS  PubMed  Google Scholar 

  45. Baass, P. C., Di Guglielmo, G. M., Authier, F., Posner, B. I. & Bergeron, J. J. M. Compartmentalized signal transduction by receptor tyrosine kinases. Trends Cell Biol. 5, 465–470 (1995).

    CAS  PubMed  Google Scholar 

  46. Grimes, M. L. et al. Endocytosis of activated TrkA: evidence that nerve growth factor induces formation of signaling endosomes. J. Neurosci. 16, 7950–7964 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Faure, R., Baquiran, G., Bergeron, J. J. & Posner, B. I. The dephosphorylation of insulin and epidermal growth factor receptors. Role of endosome-associated phosphotyrosine phosphatase(s). J. Biol. Chem. 267, 11215–11221 (1992).

    CAS  PubMed  Google Scholar 

  48. Pitcher, J. A., Payne, E. S., Csortos, C., DePaoli, R. A. & Lefkowitz, R. J. The G-protein-coupled receptor phosphatase: a protein phosphatase type 2A with a distinct subcellular distribution and substrate specificity. Proc. Natl Acad. Sci. USA 92, 8343–8347 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Grady, E. F. et al. Delineation of the endocytic pathway of substance P and its seven-transmembrane domain NK1 receptor. Mol. Biol. Cell 6, 509–524 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Law, P. Y., Hom, D. S. & Loh, H. H. Down-regulation of opiate receptor in neuroblastoma x glioma NG108-15 hybrid cells. Chloroquine promotes accumulation of tritiated enkephalin in the lysosomes. J. Biol. Chem. 259, 4096–4104 (1984).

    CAS  PubMed  Google Scholar 

  51. Gaudriault, G., Nouel, D., Dal Farra, C., Beaudet, A. & Vincent, J. P. Receptor-induced internalization of selective peptidic μ and δ opioid ligands. J. Biol. Chem. 272, 2880–2888 (1997).

    CAS  PubMed  Google Scholar 

  52. Futter, C. E., Collinson, L. M., Backer, J. M. & Hopkins, C. R. Human VPS34 is required for internal vesicle formation within multivesicular endosomes. J. Cell Biol. 155, 1251–1264 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lloyd, T. E. et al. Hrs regulates endosome membrane invagination and tyrosine kinase receptor signaling in Drosophila. Cell 108, 261–269 (2002).

    CAS  PubMed  Google Scholar 

  54. Di Gugliemo, G. M., Baass, P. C., Ou, W.-J., Posner, B. & Bergeron, J. J. M. Compartmentalization of SHC, GRB2 and mSOS, and hyperphosphorylation of Raf-1 by EGF but not insulin in liver parenchyma. EMBO J. 13, 4269–4277 (1994).This study rigorously showed the presence of RTK signalling complexes in endosomes using biochemical techniques.

    Google Scholar 

  55. Sorkin, A., McClure, M., Huang, F. & Carter, R. Interaction of EGF receptor and Grb2 in living cells visualized by fluorescence resonance energy transfer (FRET) microscopy. Curr. Biol. 10, 1395–1398 (2000).

    CAS  PubMed  Google Scholar 

  56. Oksvold, M. P., Skarpen, E., Wierod, L., Paulsen, R. E. & Huitfeldt, H. S. Re-localization of activated EGF receptor and its signal transducers to multivesicular compartments downstream of early endosomes in response to EGF. Eur. J. Cell Biol. 80, 285–294 (2001).

    CAS  PubMed  Google Scholar 

  57. Joneson, T. & Bar-Sagi, D. Ras effectors and their role in mitogenesis and oncogenesis. J. Mol. Med. 75, 587–593 (1997).

    CAS  PubMed  Google Scholar 

  58. Rizzo, M. A., Shome, K., Watkins, S. C. & Romero, G. The recruitment of Raf-1 to membranes is mediated by direct interaction with phosphatidic acid and is independent of association with Ras. J. Biol. Chem. 275, 23911–23918 (2000).

    CAS  PubMed  Google Scholar 

  59. Rizzo, M. A., Kraft, C. A., Watkins, S. C., Levitan, E. S. & Romero, G. Agonist-dependent traffic of raft-associated Ras and Raf-1 is required for activation of the mitogen-activated protein kinase cascade. J. Biol. Chem. 276, 34928–34933 (2001).

    CAS  PubMed  Google Scholar 

  60. Jiang, X. & Sorkin, A. Coordinated trafficking of Grb2 and Ras during EGF receptor endocytosis visualized in living cells. Mol. Biol. Cell 13, 1522–1535 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Choy, E. et al. Endomembrane trafficking of ras: the CAAX motif targets proteins to the ER and Golgi. Cell 98, 69–80 (1999).

    CAS  PubMed  Google Scholar 

  62. Pol, A., Calvo, M. & Enrich, C. Isolated endosomes from quiescent rat liver contain the signal transduction machinery. Differential distribution of activated Raf-1 and Mek in the endocytic compartment. FEBS Lett. 441, 34–38 (1998).

    CAS  PubMed  Google Scholar 

  63. Barbieri, M. A. et al. Epidermal growth factor and membrane trafficking. EGF receptor activation of endocytosis requires Rab5a. J. Cell Biol. 151, 539–550 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Haugh, J. M., Schooler, K., Wells, A., Wiley, H. S. & Lauffenburger, D. A. Effect of epidermal growth factor receptor internalization on regulation of the phospholipase C-γ1 signaling pathway. J. Biol. Chem. 274, 8958–8965 (1999).

    CAS  PubMed  Google Scholar 

  65. Haugh, J. M. & Meyer, T. Active EGF receptors have limited access to PtdIns(4,5)P2 in endosomes: implications for phospholipase C and PI3-kinase signaling. J. Cell Sci. 115, 303–310 (2002).

    CAS  PubMed  Google Scholar 

  66. Kranenburg, O., Verlaan, I. & Moolenaar, W. H. Dynamin is required for the activation of mitogen-activated protein (MAP) kinase by MAP kinase kinase. J. Biol. Chem. 274, 35301–35304 (1999).

    CAS  PubMed  Google Scholar 

  67. Wunderlich, W. et al. A novel 14-kilodalton protein interacts with the mitogen-activated protein kinase scaffold MP1 on a late endosomal/lysosomal compartment. J. Cell Biol. 152, 765–776 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Wu, C., Lai, C. F. & Mobley, W. C. Nerve growth factor activates persistent Rap1 signaling in endosomes. J. Neurosci. 21, 5406–5416 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. York, R. D. et al. Rap1 mediates sustained MAP kinase activation induced by nerve growth factor. Nature 392, 622–626 (1998).

    CAS  PubMed  Google Scholar 

  70. Wedegaertner, P. B. Lipid modifications and membrane targeting of Gα. Biol. Signals Recept. 7, 125–135 (1998).

    CAS  PubMed  Google Scholar 

  71. Gaidarov, I., Krupnick, J. G., Falck, J. R., Benovic, J. L. & Keen, J. H. Arrestin function in G protein-coupled receptor endocytosis requires phosphoinositide binding. EMBO J. 18, 871–881 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Perry, S. J. & Lefkowitz, R. J. Arresting developments in heptahelical receptor signaling and regulation. Trends Cell Biol. 12, 130–138 (2002).

    CAS  PubMed  Google Scholar 

  73. Oakley, R. H., Laporte, S. A., Holt, J. A., Barak, L. S. & Caron, M. G. Association of β-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitization. J. Biol. Chem. 274, 32248–32257 (1999).

    CAS  PubMed  Google Scholar 

  74. McDonald, P. H. et al. β-arrestin 2: a receptor-regulated MAPK scaffold for the activation of JNK3. Science 290, 1574–1577 (2000).This study concluded that β-arrestins have a general role as molecular scaffolds to organize MAPK modules around endocytosed GPCRs.

    CAS  PubMed  Google Scholar 

  75. Luttrell, L. M. et al. Activation and targeting of extracellular signal-regulated kinases by β-arrestin scaffolds. Proc. Natl Acad. Sci. USA 98, 2449–2454 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. DeFea, K. A. et al. β-Arrestin-dependent endocytosis of proteinase-activated receptor 2 is required for intracellular targeting of activated ERK1/2. J. Cell Biol. 148, 1267–1281 (2000).Evidence that β-arrestin controls the selectivity of GPCR signalling from the endocytic pathway by physically restricting the localization of a GPCR-activated MAPK.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Huang, E. J. & Reichardt, L. F. Neurotrophins: roles in neuronal development and function. Annu. Rev. Neurosci. 24, 677–736 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Hendry, I. A., Stockel, K., Thoenen, H. & Iversen, L. L. The retrograde axonal transport of nerve growth factor. Brain Res. 68, 103–121 (1974).

    CAS  PubMed  Google Scholar 

  79. Howe, C. L., Valletta, J. S., Rusnak, A. S. & Mobley, W. C. NGF signaling from clathrin-coated vesicles: evidence that signaling endosomes serve as a platform for the Ras–MAPK pathway. Neuron 32, 801–814 (2001).

    CAS  PubMed  Google Scholar 

  80. Riccio, A., Pierchala, B. A., Ciarallo, C. L. & Ginty, D. D. An NGF-TrkA-mediated retrograde signal to transcription factor CREB in sympathetic neurons. Science 277, 1097–1100 (1997).Evidence that neurotrophin signalling from the axon to the neural cell body is mediated by the retrograde movement of activated neurotrophin receptors after endocytosis.

    CAS  PubMed  Google Scholar 

  81. Watson, F. L. et al. Neurotrophins use the Erk5 pathway to mediate a retrograde survival response. Nature Neurosci. 4, 981–988 (2001).A study that identified a MAPK that is involved specifically in retrograde signalling by neurotrophins. This indicates how an individual growth factor might relay information that specifies both the location and nature of stimulation.

    CAS  PubMed  Google Scholar 

  82. Heerssen, H. M. & Segal, R. A. Location, location, location: a spatial view of neurotrophin signal transduction. Trends Neurosci. 25, 160–165 (2002).

    CAS  PubMed  Google Scholar 

  83. Miller, F. D. & Kaplan, D. R. On Trk for retrograde signaling. Neuron 32, 767–770 (2001).

    CAS  PubMed  Google Scholar 

  84. Whitmarsh, A. J. & Davis, R. J. Signal transduction by target-derived neurotrophins. Nature Neurosci. 4, 963–964 (2001).

    CAS  PubMed  Google Scholar 

  85. Watson, F. L. et al. Rapid nuclear responses to target-derived neurotrophins require retrograde transport of ligand–receptor complex. J. Neurosci. 19, 7889–7900 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. MacInnis, B. L. & Campenot, R. B. Retrograde support of neuronal survival without retrograde transport of nerve growth factor. Science 295, 1536–1539 (2002).

    CAS  PubMed  Google Scholar 

  87. Vieira, A. V., Lamaze, C. & Schmid, S. L. Control of EGF receptor signaling by clathrin-mediated endocytosis. Science 274, 2086–2089 (1996).The first demonstration of the role of clathrin-mediated endocytosis of RTKs in the activation of the ERK/MAPK signalling pathway.

    CAS  PubMed  Google Scholar 

  88. Zhang, Y., Moheban, D. B., Conway, B. R., Bhattacharyya, A. & Segal, R. A. Cell surface Trk receptors mediate NGF-induced survival while internalized receptors regulate NGF-induced differentiation. J. Neurosci. 20, 5671–5678 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Ceresa, B. P., Kao, A. W., Santeler, S. R. & Pessin, J. E. Inhibition of clathrin-mediated endocytosis selectively attenuates specific insulin receptor signal transduction pathways. Mol. Cell. Biol. 18, 3862–3870 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Daaka, Y. et al. Essential role for G protein-coupled receptor endocytosis in the activation of mitogen-activated protein kinase. J. Biol. Chem. 273, 685–688 (1998).

    CAS  PubMed  Google Scholar 

  91. Gutkind, J. S. The pathways connecting G protein-coupled receptors to the nucleus through divergent mitogen-activated protein kinase cascades. J. Biol. Chem. 273, 1839–1842 (1998).

    CAS  PubMed  Google Scholar 

  92. Johannessen, L. E., Ringerike, T., Molnes, J. & Madshus, I. H. Epidermal growth factor receptor efficiently activates mitogen-activated protein kinase in HeLa cells and hep2 cells conditionally defective in clathrin-dependent endocytosis. Exp. Cell Res. 260, 136–145 (2000).

    CAS  PubMed  Google Scholar 

  93. DeGraff, J. L., Gagnon, A. W., Benovic, J. L. & Orsini, M. J. Role of arrestins in endocytosis and signaling of α2-adrenergic receptor subtypes. J. Biol. Chem. 274, 11253–11259 (1999).

    CAS  PubMed  Google Scholar 

  94. Whistler, J. L. & von Zastrow, M. Dissociation of functional roles of dynamin in receptor-mediated endocytosis and mitogenic signal transduction. J. Biol. Chem. 274, 24575–24578 (1999).

    CAS  PubMed  Google Scholar 

  95. Daub, H., Weiss, F. U., Wallasch, C. & Ullrich, A. Role of transactivation of the EGF receptor in signalling by G-protein-coupled receptors. Nature 379, 557–560 (1996).

    CAS  PubMed  Google Scholar 

  96. Pierce, K. L., Maudsley, S., Daaka, Y., Luttrell, L. M. & Lefkowitz, R. J. Role of endocytosis in the activation of the extracellular signal-regulated kinase cascade by sequestering and nonsequestering G protein-coupled receptors. Proc. Natl Acad. Sci. USA 97, 1489–1494 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Luttrell, L. M. et al. β-arrestin-dependent formation of β2 adrenergic receptor–Src protein kinase complexes. Science 283, 655–661 (1999).Evidence that β-arrestins function as a scaffold that links GPCRs to activation of a non-receptor tyrosine kinase.

    CAS  PubMed  Google Scholar 

  98. McDonald, P. H. & Lefkowitz, R. J. β-arrestins: new roles in regulating heptahelical receptors' functions. Cell Signal. 13, 683–689 (2001).

    CAS  PubMed  Google Scholar 

  99. Cao, T. T., Mays, R. W. & von Zastrow, M. Regulated endocytosis of G-protein-coupled receptors by a biochemically and functionally disctinct subpopulation of clathrin-coated pits. J. Biol. Chem. 273, 24592–24602 (1998).

    CAS  PubMed  Google Scholar 

  100. Scott, M. G., Benmerah, A., Muntaner, O. & Marullo, S. Recruitment of activated G protein-coupled receptors to pre-existing clathrin-coated pits in living cells. J. Biol. Chem. 277, 3552–3559 (2002).

    CAS  PubMed  Google Scholar 

  101. Santini, F., Gaidarov, I. & Keen, J. H. G protein-coupled receptor/arrestin3 modulation of the endocytic machinery. J. Cell Biol. 156, 665–676 (2002).This study provides evidence that activated GPCRs, through interaction with arrestins, can change the properties of clathrin-coated pits through which they are endocytosed.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Connolly, J. L., Green, S. A. & Greene, L. A. Comparison of rapid changes in surface morphology and coated pit formation of PC12 cells in response to nerve growth factor, epidermal growth factor, and dibutyryl cyclic AMP. J. Cell Biol. 98, 457–465 (1984).

    CAS  PubMed  Google Scholar 

  103. Beattie, E. C., Howe, C. L., Wilde, A., Brodsky, F. M. & Mobley, W. C. NGF signals through TrkA to increase clathrin at the plasma membrane and enhance clathrin-mediated membrane trafficking. J. Neurosci. 20, 7325–7333 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Ahn, S. et al. c-Src dependent tyrosine phosphorylation regulates dynamin self-assembly and receptor-mediated endocytosis. J. Biol. Chem. 2002 May 13; [epub ahead of print].

  105. Barbieri, M. A. et al. Phosphatidylinositol-4-phosphate 5-kinase-1β is essential for epidermal growth factor receptor-mediated endocytosis. J. Biol. Chem. 276, 47212–47216 (2001).

    CAS  PubMed  Google Scholar 

  106. Wiley, H. S. Anomalous binding of epidermal growth factor to A431 cells is due to the effect of high receptor densities and a saturable endocytic system. J. Cell Biol. 107, 801–810 (1988).

    CAS  PubMed  Google Scholar 

  107. Warren, R. A., Green, F. A., Stenberg, P. E. & Enns, C. A. Distinct saturable pathways for the endocytosis of different tyrosine motifs. J. Biol. Chem. 273, 17056–17063 (1998).

    CAS  PubMed  Google Scholar 

  108. Confalonieri, S., Salcini, A. E., Puri, C., Tacchetti, C. & Di Fiore, P. P. Tyrosine phosphorylation of Eps15 is required for ligand-regulated, but not constitutive, endocytosis. J. Cell Biol. 150, 905–912 (2000).This study highlights an interesting example of the specific role of Eps15, a component of clathrin-coated pits, in the growth-factor-induced endocytosis of EGF receptors.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Tall, G. G., Barbieri, M. A., Stahl, P. D. & Horazdovsky, B. F. Ras-activated endocytosis is mediated by the Rab5 guanine nucleotide exchange activity of RIN1. Dev. Cell 1, 73–82 (2001).

    CAS  PubMed  Google Scholar 

  110. Lanzetti, L. et al. The Eps8 protein coordinates EGF receptor signalling through Rac and trafficking through Rab5. Nature 408, 374–377 (2000).

    CAS  PubMed  Google Scholar 

  111. Hicke, L. A new ticket for entry into budding vesicles — ubiquitin. Cell 106, 527–530 (2001).

    CAS  PubMed  Google Scholar 

  112. van Delft, S., Govers, R., Strous, G. J., Verkleij, A. J. & van Bergen en Henegouwen, P. M. P. Epidermal growth factor induces ubiquitination of Eps15. J. Biol. Chem. 272, 14013–14016 (1997).

    CAS  PubMed  Google Scholar 

  113. Levkowitz, G. et al. Ubiquitin ligase activity and tyrosine phosphorylation underlie suppression of growth factor signaling by c-Cbl/Sli-1. Mol. Cell 4, 1029–1040 (1999).

    CAS  PubMed  Google Scholar 

  114. Peschard, P. et al. Mutation of the c-Cbl TKB domain binding site on the Met receptor tyrosine kinase converts it into a transforming protein. Mol. Cell 8, 995–1004 (2001).

    CAS  PubMed  Google Scholar 

  115. Hicke, L. & Riezman, H. Ubiquitination of a yeast plasma membrane receptor signals its ligand-stimulated endocytosis. Cell 84, 277–287 (1996).

    CAS  PubMed  Google Scholar 

  116. Shenoy, S. K., McDonald, P. H., Kohout, T. A. & Lefkowitz, R. J. Regulation of receptor fate by ubiquitination of activated β2-adrenergic receptor and β-arrestin. Science 294, 1307–1313 (2001).

    CAS  PubMed  Google Scholar 

  117. Hopkins, C. R. Selective membrane protein trafficking: vectorial flow and filter. Trends Biochem. Sci. 17, 27–32 (1992).

    CAS  PubMed  Google Scholar 

  118. Katzmann, D. J., Babst, M. & Emr, S. D. Ubiquitin-dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT-I Cell 106, 145–155 (2001).

    CAS  PubMed  Google Scholar 

  119. Marchese, A. & Benovic, J. L. Agonist-promoted ubiquitination of the G protein-coupled receptor CXCR4 mediates lysosomal sorting. J. Biol. Chem. 276, 45509–45512 (2001).

    CAS  PubMed  Google Scholar 

  120. Kornilova, E., Sorkina, T., Beguinot, L. & Sorkin, A. Lysosomal targeting of epidermal growth factor receptors via a kinase-dependent pathway is mediated by the receptor carboxyl-terminal residues 1022–1163. J. Biol. Chem. 271, 30340–30346 (1996).

    CAS  PubMed  Google Scholar 

  121. Zerial, M. & McBride, H. Rab proteins as membrane organizers. Nature Rev. Mol. Cell Biol. 2, 107–117 (2001).

    CAS  Google Scholar 

  122. Raiborg, C., Bache, K. G., Mehlum, A., Stang, E. & Stenmark, H. Hrs recruits clathrin to early endosomes. EMBO J. 20, 5008–5021 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Komada, M. & Kitamura, N. Growth factor-induced tyrosine phosphorylation of Hrs, a novel 115-kilodalton protein with a structurally conserved putative zinc finger domain. Mol. Cell. Biol. 15, 6213–6221 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Bishop, N., Horman, A. & Woodman, P. Mammalian class E vps proteins recognize ubiquitin and act in the removal of endosomal protein–ubiquitin conjugates. J. Cell Biol. 157, 91–102 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Seachrist, J. L. et al. Rab5 association with the angiotensin II type 1A receptor promotes Rab5 GTP binding and vesicular fusion. J. Biol. Chem. 277, 679–685 (2002).

    CAS  PubMed  Google Scholar 

  126. Premont, R. T. et al. β2-adrenergic receptor regulation by GIT1, a G protein-coupled receptor kinase-associated ADP ribosylation factor GTPase-activating protein. Proc. Natl Acad. Sci. USA 95, 14082–14087 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Claing, A. et al. β-arrestin-mediated ADP-ribosylation factor 6 activation and β2-adrenergic receptor endocytosis. J. Biol. Chem. 276, 42509–42513 (2001).

    CAS  PubMed  Google Scholar 

  128. Wouters, F. S., Verveer, P. J. & Bastiaens, P. I. Imaging biochemistry inside cells. Trends Cell Biol. 11, 203–211 (2001).

    CAS  PubMed  Google Scholar 

  129. Wouters, F. S. & Bastiaens, P. I. Fluorescence lifetime imaging of receptor tyrosine kinase activity in cells. Curr. Biol. 9, 1127–1130 (1999).

    CAS  PubMed  Google Scholar 

  130. Mochizuki, N. et al. Spatio-temporal images of growth-factor-induced activation of Ras and Rap1. Nature 411, 1065–1068 (2001).

    CAS  PubMed  Google Scholar 

  131. Kurokawa, K. et al. A pair of fluorescent resonance energy transfer-based probes for tyrosine phosphorylation of the CrkII adaptor protein in vivo. J. Biol. Chem. 276, 31305–31310 (2001).

    CAS  PubMed  Google Scholar 

  132. Ting, A. Y., Kain, K. H., Klemke, R. L. & Tsien, R. Y. Genetically encoded fluorescent reporters of protein tyrosine kinase activities in living cells. Proc. Natl Acad. Sci. USA 98, 15003–15008 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Overton, M. C. & Blumer, K. J. G-protein-coupled receptors function as oligomers in vivo. Curr. Biol. 10, 341–344 (2000).

    CAS  PubMed  Google Scholar 

  134. Rocheville, M. et al. Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. J. Biol. Chem. 275, 7862–7869 (2000).

    CAS  PubMed  Google Scholar 

  135. Janetopoulos, C., Jin, T. & Devreotes, P. Receptor-mediated activation of heterotrimeric G-proteins in living cells. Science 291, 2408–2411 (2001).

    CAS  PubMed  Google Scholar 

  136. Barak, L. S., Ferguson, S. S., Zhang, J. & Caron, M. G. A β-arrestin/green fluorescent protein biosensor for detecting G protein-coupled receptor activation. J. Biol. Chem. 272, 27497–27500 (1997).

    CAS  PubMed  Google Scholar 

  137. Schulz, R., Wehmeyer, A. & Schulz, K. Opioid receptor types selectively cointernalize with G protein-coupled receptor kinases 2 and 3. J. Pharmacol. Exp. Ther. 300, 376–384 (2002).

    CAS  PubMed  Google Scholar 

  138. Carman, C. V. & Benovic, J. L. G-protein-coupled receptors: turn-ons and turn-offs. Curr. Opin. Neurobiol. 8, 335–344 (1998).

    CAS  PubMed  Google Scholar 

  139. Innamorati, G., Sadeghi, H. M., Tran, N. T. & Birnbaumer, M. A serine cluster prevents recycling of the V2 vasopressin receptor. Proc. Natl Acad. Sci. USA 95, 2222–2226 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Tsao, P. I. & von Zastrow, M. Type-specific sorting of G protein-coupled receptors after endocytosis. J. Biol. Chem. 275, 11130–11140 (2000).

    CAS  PubMed  Google Scholar 

  141. Cao, T. T., Deacon, H. W., Reczek, D., Bretscher, A. & von Zastrow, M. A kinase-regulated PDZ-domain interaction controls endocytic sorting of the β2-adrenergic receptor. Nature 401, 286–290 (1999).

    CAS  PubMed  Google Scholar 

  142. Laporte, S. A., Miller, W. E., Kim, K. M. & Caron, M. G. β-Arrestin/AP-2 interaction in G protein-coupled receptor internalization. Identification of a β-arrestin binding site in β2-adaptin. J. Biol. Chem. 277, 9247–9254 (2002).

    CAS  PubMed  Google Scholar 

  143. Artavanis-Tsakonas, S., Rand, M. D. & Lake, R. J. Notch signaling: cell fate control and signal integration in development. Science 284, 770–776 (1999).

    CAS  PubMed  Google Scholar 

  144. DeFea, K. A. et al. The proliferative and antiapoptotic effects of substance P are facilitated by formation of a β-arrestin-dependent scaffolding complex. Proc. Natl Acad. Sci. USA 97, 11086–11091 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Chen, H. et al. Epsin is an EH-domain-binding protein implicated in clathrin-mediated endocytosis. Nature 394, 793–797 (1998).

    CAS  PubMed  Google Scholar 

  146. Nakashima, S. et al. Small G protein Ral and its downstream molecules regulate endocytosis of EGF and insulin receptors. EMBO J. 18, 3629–3642 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Kurten, R. C., Cadena, D. L. & Gill, G. N. Enhanced degradation of EGF receptors by a sorting nexin, SNX-1. Science 272, 1008–1010 (1996).

    CAS  PubMed  Google Scholar 

  148. Takata, H., Kato, M., Denda, K. & Kitamura, N. A hrs binding protein having a Src homology 3 domain is involved in intracellular degradation of growth factors and their receptors. Genes Cells 5, 57–69 (2000).

    CAS  PubMed  Google Scholar 

  149. Xu, Y., Hortsman, H., Seet, L., Wong, S. H. & Hong, W. SNX3 regulates endosomal function through its PX-domain-mediated interaction with PtdIns3P. Nature Cell Biol. 3, 658–666 (2001).

    CAS  PubMed  Google Scholar 

  150. Burke, P., Schooler, K. & Wiley, H. S. Regulation of epidermal growth factor receptor signaling by endocytosis and intracellular trafficking. Mol. Biol. Cell 12, 1897–1910 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Kelly, K. L. & Ruderman, N. B. Insulin-stimulated phosphatidylinositol 3-kinase. Association with a 185-kDa tyrosine-phosphorylated protein (IRS-1) and localization in a low density membrane vesicle. J. Biol. Chem. 268, 4391–4398 (1993).

    CAS  PubMed  Google Scholar 

  152. Wang, Z., Tung, P. S. & Moran, M. F. Association of p120 ras GAP with endocytic components and colocalization with epidermal growth factor (EGF) receptor in response to EGF stimulation. Cell Growth Differ. 7, 123–133 (1996).

    CAS  PubMed  Google Scholar 

  153. Wang, X. J., Liao, H. J., Chattopadhyay, A. & Carpenter, G. EGF-dependent translocation of green fluorescent protein-tagged PLC-γ1 to the plasma membrane and endosomes. Exp. Cell Res. 267, 28–36 (2001).

    CAS  PubMed  Google Scholar 

  154. Matsuda, M. et al. Real time fluorescence imaging of PLCγ translocation and its interaction with the epidermal growth factor receptor. J. Cell Biol. 153, 599–612 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Kapeller, R., Chakrabarti, R., Cantley, L., Fay, F. & Corvera, S. Internalization of activated platelet-derived growth factor receptor-phosphatidylinositol-3′ kinase complexes: potential interactions with the microtubule cytoskeleton. Mol. Cell. Biol. 13, 6052–6063 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Zheng, B. et al. RGS-PX1, a GAP for GαS and sorting nexin in vesicular trafficking. Science 294, 1939–1942 (2001).

    CAS  PubMed  Google Scholar 

  157. Kaplan, K. B., Swedlow, J. R., Varmus, H. E. & Morgan, D. O. Association of p60c-src with endosomal membranes in mammalian fibroblasts. J. Cell Biol. 118, 321–333 (1992).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A.S. was supported by grants from the National Cancer Institute, National Institute on Drug Abuse, National Science Foundation and the American Cancer Society. M.v.Z. was supported by grants from the National Institute on Drug Abuse.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alexander Sorkin.

Related links

Related links

DATABASES

FlyBase

RN-tre

Delta

dor

dpp

engrailed

Neuralized

Notch

SOS

Wg

LocusLink

B-Raf

C3G

CrkL

EGF

EGF receptor

ERK5

Gab2

PI3K

Rab5

Rap1

RIN1

TrkA

Swiss-Prot

β-adrenergic receptor

c-Raf-1

Grb2

MEK

Shc

Glossary

SH2 DOMAIN

(Src-homology-2 domain). A protein motif that recognizes and binds tyrosine-phosphorylated sequences, and thereby has a key role in relaying cascades of signal transduction.

PTB DOMAIN

(Phosphotyrosine-binding domain). Also known as a phosphotyrosine interaction domain (PID), this motif is similar to an SH2 domain in that it binds tyrosine-phosphorylated sequences, and thereby mediates signal transduction.

GUANINE-NUCLEOTIDE EXCHANGE FACTOR

(GEF). A protein that facilitates the release of GDP from, and the binding of the more abundant GTP to, small GTP-binding proteins.

ENDOCYTOSIS

The uptake of extracellular materials by cells. The plasma membrane invaginates and vesicles that contain endocytosed molecules and plasma membrane components pinch off.

CLATHRIN

A large protein, which polymerizes into a triskelion, and comprises three heavy chains and three light chains. Triskelions assemble into polyhedral lattices to form clathrin coats.

MICROPINOCYTOSIS

An actin-dependent process, in which the pinosomes that are formed are very small and can only be visualized using the electron microscope.

LYSOSOME

A membrane-bounded organelle with a low internal pH (4–5) that contains hydrolytic enzymes and that is the site of the degradation of proteins in both the biosynthetic and the endocytic pathways.

MULTIVESICULAR BODIES

Endosomal intermediates in which small membrane vesicles are enclosed within a limiting membrane. The internal vesicles are thought to form by invagination and budding from the limiting membrane.

GREEN FLUORESCENT PROTEIN

(GFP). An autofluorescent protein that was originally isolated from the jellyfish Aequorea victoria. Can be genetically conjugated with proteins to make them fluorescent. The most widely used mutant, enhanced (E)GFP, has an emission maximum at 510 nm.

RAB PROTEINS

These form the largest subfamily of small GTPases of the Ras superfamily. They regulate budding, tethering, fusion and motility at various sites within cells.

IMAGINAL DISCS

Sac-like structures that are present in the larvae and are composed of cells destined to form the adult cuticular structures. They are named after the adult part that they make; for example, wing, leg, eye-antennal or genital.

RING

A cysteine-rich 'RING'-finger domain of 40–60 amino acids (also called the C3HC4 Zn finger), which binds two atoms of Zn and might mediate protein–protein interactions. Most RING-finger proteins have been shown to bind DNA.

SUBSTANCE P

Substance P is an 11-amino-acid tachykinin peptide neurotransmitter that binds preferentially to the NK1 receptor. A second tachykinin (NKA) also has high affinity for this receptor, which indicates that to refer to the NK1 receptor as 'the substance-P receptor' could be misleading.

ADAPTOR PROTEIN

A protein that augments cellular responses by recruiting other proteins to a complex. Adaptor proteins usually contain several protein–protein interaction domains.

PC12 CELLS

A clonal line of rat adrenal pheochromocytoma cells that responds to nerve growth factor and can synthesize, store and secrete catecholamines, much like sympathetic neurons. PC12 cells contain small, clear synaptic-like vesicles and larger, dense core granules.

PALMITOYL

The common name for hexadecanoyl, the acyl group that is derived from palmitic acid.

MYRISTOYL

The common name for tetradecanoyl, the acyl group that is derived from myristic acid.

FLUORESCENCE RESONANCE ENERGY TRANSFER

(FRET). A technique in which a fluorophore donor molecule is excited and transfers the energy of an adsorbed photon to an acceptor molecule.

ENDOTHELIAL CELLS

Flattened cells that grow in a single layer and line blood vessels.

CREB

A transcription factor that functions in glucose homeostasis and growth-factor-dependent cell survival, and has also been implicated in learning and memory.

MICROTUBULE

A hollow tube, 25 nm in diameter, that is formed by the lateral association of 13 protofilaments, which are themselves polymers of α- and β-tubulin subunits.

EPS15

(Epidermal-growth-factor-receptor pathway substrate clone 15). A mammalian protein that is required for budding of clathrin-coated vesicles during endocytosis.

GTPASE ACTIVATING PROTEINS

(GAPs). Proteins that inactivate small GTP-binding proteins, such as Ras family members, by increasing their rate of GTP hydrolysis.

E3 UBIQUITIN PROTEIN LIGASE

The third enzyme in a series — the first two are designated E1 and E2 — that are responsible for ubiquitylation of target proteins. E3 enzymes provide platforms for binding E2 enzymes and specific substrates, thereby coordinating ubiquitylation of the selected substrates.

UBIQUITYLATION

The attachment of the protein ubiquitin to lysine residues of other molecules, often as a tag for their rapid cellular degradation.

FYVE DOMAIN

A protein motif that binds phosphatidylinositol 3-phosphate and thereby mediates docking of cytosolic proteins to the membrane.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sorkin, A., von Zastrow, M. Signal transduction and endocytosis: close encounters of many kinds. Nat Rev Mol Cell Biol 3, 600–614 (2002). https://doi.org/10.1038/nrm883

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm883

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