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:

Endocytosis in the context-dependent regulation of individual and collective cell properties

Abstract

Endocytosis allows cells to transport particles and molecules across the plasma membrane. In addition, it is involved in the termination of signalling through receptor downmodulation and degradation. This traditional outlook has been substantially modified in recent years by discoveries that endocytosis and subsequent trafficking routes have a profound impact on the positive regulation and propagation of signals, being key for the spatiotemporal regulation of signal transmission in cells. Accordingly, endocytosis and membrane trafficking regulate virtually every aspect of cell physiology and are frequently subverted in pathological conditions. Two key aspects of endocytic control over signalling are coming into focus: context-dependency and long-range effects. First, endocytic-regulated outputs are not stereotyped but heavily dependent on the cell-specific regulation of endocytic networks. Second, endocytic regulation has an impact not only on individual cells but also on the behaviour of cellular collectives. Herein, we will discuss recent advancements in these areas, highlighting how endocytic trafficking impacts complex cell properties, including cell polarity and collective cell migration, and the relevance of these mechanisms to disease, in particular cancer.

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

Fig. 1: Heterogeneity of clathrin-coated pits.
Fig. 2: NCE mechanisms and cellular contexts.
Fig. 3: Membrane trafficking in apical–basal cell polarity.
Fig. 4: Membrane trafficking in apical lumen formation.
Fig. 5: Endocytosis and actomyosin in the control of cell shape and tissue elongation.
Fig. 6: Endomembranes in the dynamics of collective motion.

Similar content being viewed by others

References

  1. Sigismund, S. et al. Endocytosis and signaling: cell logistics shape the eukaryotic cell plan. Physiol. Rev. 92, 273–366 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Thottacherry, J. J., Sathe, M., Prabhakara, C. & Mayor, S. Spoiled for choice: diverse endocytic pathways function at the cell surface. Annu. Rev. Cell Dev. Biol. 35, 55–84 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Goh, L. K. & Sorkin, A. Endocytosis of receptor tyrosine kinases. Cold Spring Harb. Perspect. Biol. 5, a017459 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Irannejad, R., Tsvetanova, N. G., Lobingier, B. T. & von Zastrow, M. Effects of endocytosis on receptor-mediated signaling. Curr. Opin. Cell Biol. 35, 137–143 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Villasenor, R., Kalaidzidis, Y. & Zerial, M. Signal processing by the endosomal system. Curr. Opin. Cell Biol. 39, 53–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Lanzetti, L. & Di Fiore, P. P. Behind the scenes: endo/exocytosis in the acquisition of metastatic traits. Cancer Res. 77, 1813–1817 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Wilson, B. J., Allen, J. L. & Caswell, P. T. Vesicle trafficking pathways that direct cell migration in 3D matrices and in vivo. Traffic 19, 899–909 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Disanza, A., Frittoli, E., Palamidessi, A. & Scita, G. Endocytosis and spatial restriction of cell signaling. Mol. Oncol. 3, 280–296 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Eaton, S. & Martin-Belmonte, F. Cargo sorting in the endocytic pathway: a key regulator of cell polarity and tissue dynamics. Cold Spring Harb. Perspect. Biol. 6, a016899 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sigismund, S. & Scita, G. The ‘endocytic matrix reloaded’ and its impact on the plasticity of migratory strategies. Curr. Opin. Cell Biol. 54, 9–17 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Katsuno-Kambe, H. & Yap, A. S. Endocytosis, cadherins and tissue dynamics. Traffic 21, 268–273 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Ladoux, B., Mege, R. M. & Trepat, X. Front-rear polarization by mechanical cues: from single cells to tissues. Trends Cell Biol. 26, 420–433 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Gonzalez-Gaitan, M. & Julicher, F. The role of endocytosis during morphogenetic signaling. Cold Spring Harb. Perspect. Biol. 6, a016881 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Johannes, L., Parton, R. G., Bassereau, P. & Mayor, S. Building endocytic pits without clathrin. Nat. Rev. Mol. Cell Biol. 16, 311–321 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Ferreira, A. P. A. & Boucrot, E. Mechanisms of carrier formation during clathrin-independent endocytosis. Trends Cell Biol. 28, 188–200 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Azarnia Tehran, D., Lopez-Hernandez, T. & Maritzen, T. Endocytic adaptor proteins in health and disease: lessons from model organisms and human mutations. Cells 8, 1345 (2019).

    Article  PubMed Central  CAS  Google Scholar 

  17. Khan, I. & Steeg, P. S. Endocytosis: a pivotal pathway for regulating metastasis. Br. J. Cancer 124, 66–75 (2021).

    Article  CAS  PubMed  Google Scholar 

  18. Martello, A., Platt, F. M. & Eden, E. R. Staying in touch with the endocytic network: The importance of contacts for cholesterol transport. Traffic 21, 354–363 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wu, H., Carvalho, P. & Voeltz, G. K. Here, there, and everywhere: the importance of ER membrane contact sites. Science 361, eaan5835 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Scorrano, L. et al. Coming together to define membrane contact sites. Nat. Commun. 10, 1287 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Kaempf, N. & Maritzen, T. Safeguards of neurotransmission: endocytic adaptors as regulators of synaptic vesicle composition and function. Front. Cell Neurosci. 11, 320 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Saheki, Y. & De Camilli, P. Synaptic vesicle endocytosis. Cold Spring Harb. Perspect. Biol. 4, a005645 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Soykan, T., Maritzen, T. & Haucke, V. Modes and mechanisms of synaptic vesicle recycling. Curr. Opin. Neurobiol. 39, 17–23 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Moreno-Layseca, P., Icha, J., Hamidi, H. & Ivaska, J. Integrin trafficking in cells and tissues. Nat. Cell Biol. 21, 122–132 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bruser, L. & Bogdan, S. Adherens junctions on the move-membrane trafficking of E-Cadherin. Cold Spring Harb. Perspect Biol. 9, a029140 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Nino, C. A., Sala, S. & Polo, S. When ubiquitin meets E-cadherin: plasticity of the epithelial cellular barrier. Semin. Cell Dev. Biol. 93, 136–144 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Lampe, M., Vassilopoulos, S. & Merrifield, C. Clathrin coated pits, plaques and adhesion. J. Struct. Biol. 196, 48–56 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. De Deyne, P. G. et al. The vitronectin receptor associates with clathrin-coated membrane domains via the cytoplasmic domain of its beta5 subunit. J. Cell Sci. 111, 2729–2740 (1998).

    Article  PubMed  Google Scholar 

  29. Heuser, J. Three-dimensional visualization of coated vesicle formation in fibroblasts. J. Cell Biol. 84, 560–583 (1980).

    Article  CAS  PubMed  Google Scholar 

  30. Maupin, P. & Pollard, T. D. Improved preservation and staining of HeLa cell actin filaments, clathrin-coated membranes, and other cytoplasmic structures by tannic acid-glutaraldehyde-saponin fixation. J. Cell Biol. 96, 51–62 (1983).

    Article  CAS  PubMed  Google Scholar 

  31. Saffarian, S., Cocucci, E. & Kirchhausen, T. Distinct dynamics of endocytic clathrin-coated pits and coated plaques. PLoS Biol. 7, e1000191 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Baschieri, F. et al. Frustrated endocytosis controls contractility-independent mechanotransduction at clathrin-coated structures. Nat. Commun. 9, 3825 (2018). This paper describes a function for clathrin-coated plaques as contractility-independent mechanosensitive structures that assemble with increasing substrate rigidity and that serve as platforms for receptor tyrosine kinase signalling.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Lock, J. G. et al. Clathrin-containing adhesion complexes. J. Cell Biol. 218, 2086–2095 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kirchhausen, T., Owen, D. & Harrison, S. C. Molecular structure, function, and dynamics of clathrin-mediated membrane traffic. Cold Spring Harb. Perspect. Biol. 6, a016725 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Mettlen, M., Chen, P. H., Srinivasan, S., Danuser, G. & Schmid, S. L. Regulation of Clathrin-Mediated Endocytosis. Annu. Rev. Biochem. 87, 871–896 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Antonny, B. et al. Membrane fission by dynamin: what we know and what we need to know. EMBO J. 35, 2270–2284 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ramachandran, R. & Schmid, S. L. The dynamin superfamily. Curr. Biol. 28, R411–R416 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Mettlen, M. et al. Endocytic accessory proteins are functionally distinguished by their differential effects on the maturation of clathrin-coated pits. Mol. Biol. Cell 20, 3251–3260 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kaksonen, M. & Roux, A. Mechanisms of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 19, 313–326 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Dambournet, D. et al. Genome-edited human stem cells expressing fluorescently labeled endocytic markers allow quantitative analysis of clathrin-mediated endocytosis during differentiation. J. Cell Biol. 217, 3301–3311 (2018). Using genome-edited human embryonic stem cells to derive isogenic fibroblasts and neuronal progenitors, the authors show that the levels of expression of the endocytic adaptor AP2 are cell context-regulated and that this impinges on CME dynamics.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hanyaloglu, A. C. & von Zastrow, M. Regulation of GPCRs by endocytic membrane trafficking and its potential implications. Annu. Rev. Pharmacol. Toxicol. 48, 537–568 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Puthenveedu, M. A. & von Zastrow, M. Cargo regulates clathrin-coated pit dynamics. Cell 127, 113–124 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Henry, A. G. et al. Regulation of endocytic clathrin dynamics by cargo ubiquitination. Dev. Cell 23, 519–532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Soohoo, A. L. & Puthenveedu, M. A. Divergent modes for cargo-mediated control of clathrin-coated pit dynamics. Mol. Biol. Cell 24, 1725–1734, S1-S12 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Eichel, K., Jullie, D. & von Zastrow, M. beta-Arrestin drives MAP kinase signalling from clathrin-coated structures after GPCR dissociation. Nat. Cell Biol. 18, 303–310 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Eichel, K. et al. Catalytic activation of beta-arrestin by GPCRs. Nature 557, 381–386 (2018). This study demonstrates an additional mechanism of β-arrestin activation, which does not require a stable GPCR–β-arrestin binding and promotes the accumulation of β-arrestin in clathrin-coated pits after dissociation from the GPCR, leading to ERK signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Latorraca, N. R. et al. Molecular mechanism of GPCR-mediated arrestin activation. Nature 557, 452–456 (2018). The mechanism of receptor-mediated arrestin activation is herein investigated through atomic-level simulations, revealing that, in the absence of a receptor, arrestin frequently adopts active conformations, which may explain why arrestin may remain active also after receptor dissociation (as shown by Eichel et al.111).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Huang, F., Khvorova, A., Marshall, W. & Sorkin, A. Analysis of clathrin-mediated endocytosis of epidermal growth factor receptor by RNA interference. J. Biol. Chem. 279, 16657–16661 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Motley, A., Bright, N. A., Seaman, M. N. & Robinson, M. S. Clathrin-mediated endocytosis in AP-2-depleted cells. J. Cell Biol. 162, 909–918 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hinrichsen, L., Harborth, J., Andrees, L., Weber, K. & Ungewickell, E. J. Effect of clathrin heavy chain- and alpha-adaptin-specific small inhibitory RNAs on endocytic accessory proteins and receptor trafficking in HeLa cells. J. Biol. Chem. 278, 45160–45170 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Pascolutti, R. et al. Molecularly distinct clathrin-coated pits differentially impact EGFR fate and signaling. Cell Rep. 27, 3049–3061 e3046 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ko, G. et al. Selective high-level expression of epsin 3 in gastric parietal cells, where it is localized at endocytic sites of apical canaliculi. Proc. Natl Acad. Sci. USA 107, 21511–21516 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Schiano Lomoriello, I. et al. A self-sustaining endocytic-based loop promotes breast cancer plasticity leading to aggressiveness and pro-metastatic behavior. Nat. Commun. 11, 3020 (2020). This study shows that the endocytic protein EPN3 is an oncogene with prognostic relevance in breast cancer and that it drives breast tumorigenesis through the induction of E-cadherin endocytosis, EMT and invasive behaviour.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sen, A., Madhivanan, K., Mukherjee, D. & Aguilar, R. C. The epsin protein family: coordinators of endocytosis and signaling. Biomol. Concepts 3, 117–126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kazazic, M. et al. Epsin 1 is involved in recruitment of ubiquitinated EGF receptors into clathrin-coated pits. Traffic 10, 235–245 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Sigismund, S. et al. Clathrin-independent endocytosis of ubiquitinated cargos. Proc. Natl Acad. Sci. USA 102, 2760–2765 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chang, B. et al. Epsin is required for Dishevelled stability and Wnt signalling activation in colon cancer development. Nat. Commun. 6, 6380 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  59. Langridge, P. D. & Struhl, G. Epsin-dependent ligand endocytosis activates notch by force. Cell 171, 1383–1396.e12 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pasula, S. et al. Endothelial epsin deficiency decreases tumor growth by enhancing VEGF signaling. J. Clin. Invest. 122, 4424–4438 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Spradling, K. D., McDaniel, A. E., Lohi, J. & Pilcher, B. K. Epsin 3 is a novel extracellular matrix-induced transcript specific to wounded epithelia. J. Biol. Chem. 276, 29257–29267 (2001).

    Article  CAS  PubMed  Google Scholar 

  62. Ferguson, S. M. et al. A selective activity-dependent requirement for dynamin 1 in synaptic vesicle endocytosis. Science 316, 570–574 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Ferguson, S. M. et al. Coordinated actions of actin and BAR proteins upstream of dynamin at endocytic clathrin-coated pits. Dev. Cell 17, 811–822 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Liu, Y. W., Surka, M. C., Schroeter, T., Lukiyanchuk, V. & Schmid, S. L. Isoform and splice-variant specific functions of dynamin-2 revealed by analysis of conditional knock-out cells. Mol. Biol. Cell 19, 5347–5359 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Reis, C. R. et al. Crosstalk between Akt/GSK3beta signaling and dynamin-1 regulates clathrin-mediated endocytosis. EMBO J. 34, 2132–2146 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Srinivasan, S. et al. A noncanonical role for dynamin-1 in regulating early stages of clathrin-mediated endocytosis in non-neuronal cells. PLoS Biol. 16, e2005377 (2018). Shows that, although highly enriched in neurons, dynamin 1 is expressed in non-neuronal cells but inactivated via phosphorylation by GSK3β and that it becomes activated downstream of EGFR signalling to regulate dynamics of clathrin-coated pits.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Loerke, D. et al. Cargo and dynamin regulate clathrin-coated pit maturation. PLoS Biol. 7, e57 (2009).

    Article  PubMed  CAS  Google Scholar 

  68. Clayton, E. L. et al. Dynamin I phosphorylation by GSK3 controls activity-dependent bulk endocytosis of synaptic vesicles. Nat. Neurosci. 13, 845–851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Meng, J. Distinct functions of dynamin isoforms in tumorigenesis and their potential as therapeutic targets in cancer. Oncotarget 8, 41701–41716 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Gong, C. et al. Dynamin2 downregulation delays EGFR endocytic trafficking and promotes EGFR signaling and invasion in hepatocellular carcinoma. Am. J. Cancer Res. 5, 702–713 (2015).

    PubMed  PubMed Central  Google Scholar 

  71. Ezratty, E. J., Bertaux, C., Marcantonio, E. E. & Gundersen, G. G. Clathrin mediates integrin endocytosis for focal adhesion disassembly in migrating cells. J. Cell Biol. 187, 733–747 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Burton, K. M. et al. Dynamin 2 interacts with alpha-actinin 4 to drive tumor cell invasion. Mol. Biol. Cell 31, 439–451 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Bhave, M., Mettlen, M., Wang, X. & Schmid, S. L. Early and non-redundant functions of dynamin isoforms in clathrin-mediated endocytosis. Mol. Biol. Cell 31, 2035–2047 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Reis, C. R., Chen, P. H., Bendris, N. & Schmid, S. L. TRAIL-death receptor endocytosis and apoptosis are selectively regulated by dynamin-1 activation. Proc. Natl Acad. Sci. USA 114, 504–509 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Parton, R. G. et al. Caveolae: the FAQs. Traffic 21, 181–185 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Parton, R. G., McMahon, K. A. & Wu, Y. Caveolae: formation, dynamics, and function. Curr. Opin. Cell Biol. 65, 8–16 (2020).

    Article  CAS  PubMed  Google Scholar 

  77. Sabharanjak, S., Sharma, P., Parton, R. G. & Mayor, S. GPI-anchored proteins are delivered to recycling endosomes via a distinct cdc42-regulated, clathrin-independent pinocytic pathway. Dev. Cell 2, 411–423 (2002).

    Article  CAS  PubMed  Google Scholar 

  78. Kirkham, M. et al. Ultrastructural identification of uncoated caveolin-independent early endocytic vehicles. J. Cell Biol. 168, 465–476 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Holst, M. R. et al. Clathrin-Independent endocytosis suppresses cancer cell blebbing and invasion. Cell Rep. 20, 1893–1905 (2017).

    Article  CAS  PubMed  Google Scholar 

  80. Howes, M. T. et al. Clathrin-independent carriers form a high capacity endocytic sorting system at the leading edge of migrating cells. J. Cell Biol. 190, 675–691 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Thottacherry, J. J. et al. Mechanochemical feedback control of dynamin independent endocytosis modulates membrane tension in adherent cells. Nat. Commun. 9, 4217 (2018). This study describes a role for CLIC/GEEC endocytosis as a critical regulator of membrane tension in adherent cells and dissects the downstream molecular mechanism, which involves vinculin as a mechanotransducer at focal adhesion sites.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Goldmann, W. H. Role of vinculin in cellular mechanotransduction. Cell Biol. Int. 40, 241–256 (2016).

    Article  PubMed  Google Scholar 

  83. del Pozo, M. A. et al. Phospho-caveolin-1 mediates integrin-regulated membrane domain internalization. Nat. Cell Biol. 7, 901–908 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Boucrot, E. et al. Endophilin marks and controls a clathrin-independent endocytic pathway. Nature 517, 460–465 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Casamento, A. & Boucrot, E. Molecular mechanism of fast endophilin-mediated endocytosis. Biochem. J. 477, 2327–2345 (2020).

    Article  PubMed  Google Scholar 

  86. Sigismund, S. et al. Clathrin-mediated internalization is essential for sustained EGFR signaling but dispensable for degradation. Dev. Cell 15, 209–219 (2008).

    Article  CAS  PubMed  Google Scholar 

  87. Caldieri, G. et al. Reticulon 3-dependent ER-PM contact sites control EGFR nonclathrin endocytosis. Science 356, 617–624 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Ghosh, S., Marrocco, I. & Yarden, Y. Roles for receptor tyrosine kinases in tumor progression and implications for cancer treatment. Adv. Cancer Res. 147, 1–57 (2020).

    Article  PubMed  Google Scholar 

  89. Schlessinger, J. Receptor tyrosine kinases: legacy of the first two decades. Cold Spring Harb. Perspect Biol. 6, a008912 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Lamaze, C. et al. Interleukin 2 receptors and detergent-resistant membrane domains define a clathrin-independent endocytic pathway. Mol. Cell 7, 661–671 (2001).

    Article  CAS  PubMed  Google Scholar 

  91. Hemar, A. et al. Endocytosis of interleukin 2 receptors in human T lymphocytes: distinct intracellular localization and fate of the receptor alpha, beta, and gamma chains. J. Cell Biol. 129, 55–64 (1995).

    Article  CAS  PubMed  Google Scholar 

  92. Sehat, B., Andersson, S., Girnita, L. & Larsson, O. Identification of c-Cbl as a new ligase for insulin-like growth factor-I receptor with distinct roles from Mdm2 in receptor ubiquitination and endocytosis. Cancer Res. 68, 5669–5677 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Salani, B. et al. IGF-IR internalizes with Caveolin-1 and PTRF/Cavin in HaCat cells. PLoS One 5, e14157 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. De Donatis, A. et al. Proliferation versus migration in platelet-derived growth factor signaling: the key role of endocytosis. J. Biol. Chem. 283, 19948–19956 (2008).

    Article  PubMed  CAS  Google Scholar 

  95. Jastrzebski, K. et al. Multiple routes of endocytic internalization of PDGFRbeta contribute to PDGF-induced STAT3 signaling. J. Cell Sci. 130, 577–589 (2017).

    CAS  PubMed  Google Scholar 

  96. Sadowski, L. et al. Dynamin inhibitors impair endocytosis and mitogenic signaling of PDGF. Traffic 14, 725–736 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Basagiannis, D. et al. VEGF induces signalling and angiogenesis by directing VEGFR2 internalisation through macropinocytosis. J. Cell Sci. 129, 4091–4104 (2016).

    CAS  PubMed  Google Scholar 

  98. Genet, G. et al. Endophilin-A2 dependent VEGFR2 endocytosis promotes sprouting angiogenesis. Nat. Commun. 10, 2350 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Nakayama, M. et al. Spatial regulation of VEGF receptor endocytosis in angiogenesis. Nat. Cell Biol. 15, 249–260 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Sawamiphak, S. et al. Ephrin-B2 regulates VEGFR2 function in developmental and tumour angiogenesis. Nature 465, 487–491 (2010).

    Article  CAS  PubMed  Google Scholar 

  101. Marques, P. E., Grinstein, S. & Freeman, S. A. SnapShot:Macropinocytosis. Cell 169, 766–766.e1 (2017).

    Article  CAS  PubMed  Google Scholar 

  102. Lin, X. P., Mintern, J. D. & Gleeson, P. A. Macropinocytosis in different cell types: similarities and differences. Membranes 10, 177 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  103. Bar-Sagi, D. & Feramisco, J. R. Induction of membrane ruffling and fluid-phase pinocytosis in quiescent fibroblasts by ras proteins. Science 233, 1061–1068 (1986).

    Article  CAS  PubMed  Google Scholar 

  104. Porat-Shliom, N., Kloog, Y. & Donaldson, J. G. A unique platform for H-Ras signaling involving clathrin-independent endocytosis. Mol. Biol. Cell 19, 765–775 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Walsh, A. B. & Bar-Sagi, D. Differential activation of the Rac pathway by Ha-Ras and K-Ras. J. Biol. Chem. 276, 15609–15615 (2001).

    Article  CAS  PubMed  Google Scholar 

  106. Recouvreux, M. V. & Commisso, C. Macropinocytosis: a metabolic adaptation to nutrient stress in cancer. Front. Endocrinol. 8, 261 (2017).

    Article  Google Scholar 

  107. Kim, S. M. et al. PTEN deficiency and AMPK activation promote nutrient scavenging and anabolism in prostate cancer cells. Cancer Discov. 8, 866–883 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Jayashankar, V. & Edinger, A. L. Macropinocytosis confers resistance to therapies targeting cancer anabolism. Nat. Commun. 11, 1121 (2020). This study establishes necrocytosis as a mechanism of drug resistance, demonstrating its role in supplying amino acids, sugars, fatty acids, and nucleotides for biosynthesis and evidencing that it confers resistance to therapies targeting anabolic pathways in a cell context-dependent manner.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Jayashankar, V., Finicle, B. T. & Edinger, A. L. Starving PTEN-deficient prostate cancer cells thrive under nutrient stress by scavenging corpses for their supper. Mol. Cell Oncol. 5, e1472060 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Norris, A. & Grant, B. D. Endosomal microdomains: formation and function. Curr. Opin. Cell Biol. 65, 86–95 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Eichel, K. & von Zastrow, M. Subcellular organization of GPCR signaling. Trends Pharmacol. Sci. 39, 200–208 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Irannejad, R. & von Zastrow, M. GPCR signaling along the endocytic pathway. Curr. Opin. Cell Biol. 27, 109–116 (2014).

    Article  CAS  PubMed  Google Scholar 

  113. Jha, A., van Zanten, T. S., Philippe, J. M., Mayor, S. & Lecuit, T. Quantitative control of GPCR organization and signaling by endocytosis in epithelial morphogenesis. Curr. Biol. 28, 1570–1584 e1576 (2018). Shows that in the D. melanogaster embryo, the dynamic partitioning of active GPCRs at the plasma membrane or in plasma membrane invaginations by endocytosis creates platforms for RHO1 signalling and MyoII activation, which regulate epithelial morphogenesis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kerridge, S. et al. Modular activation of Rho1 by GPCR signalling imparts polarized myosin II activation during morphogenesis. Nat. Cell Biol. 18, 261–270 (2016).

    Article  CAS  PubMed  Google Scholar 

  115. Irannejad, R. et al. Functional selectivity of GPCR-directed drug action through location bias. Nat. Chem. Biol. 13, 799–806 (2017). In this study, the human β1-adrenergic receptor is shown to induce cAMP signalling from the Golgi apparatus, leading authors to propose ‘location bias’ as a new principle for achieving functional selectivity of GPCR-directed drug action.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Godbole, A., Lyga, S., Lohse, M. J. & Calebiro, D. Internalized TSH receptors en route to the TGN induce local Gs-protein signaling and gene transcription. Nat. Commun. 8, 443 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Boivin, B., Vaniotis, G., Allen, B. G. & Hebert, T. E. G protein-coupled receptors in and on the cell nucleus: a new signaling paradigm? J. Recept. Signal. Transduct. Res. 28, 15–28 (2008).

    Article  CAS  PubMed  Google Scholar 

  118. Calebiro, D. et al. Persistent cAMP-signals triggered by internalized G-protein-coupled receptors. PLoS Biol. 7, e1000172 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Inda, C. et al. Different cAMP sources are critically involved in G protein-coupled receptor CRHR1 signaling. J. Cell Biol. 214, 181–195 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Lazar, A. M. et al. G protein-regulated endocytic trafficking of adenylyl cyclase type 9. eLife 9, e58039 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Tsvetanova, N. G. & von Zastrow, M. Spatial encoding of cyclic AMP signaling specificity by GPCR endocytosis. Nat. Chem. Biol. 10, 1061–1065 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Ritter, S. L. & Hall, R. A. Fine-tuning of GPCR activity by receptor-interacting proteins. Nat. Rev. Mol. Cell Biol. 10, 819–830 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Weinberg, Z. Y. & Puthenveedu, M. A. Regulation of G protein-coupled receptor signaling by plasma membrane organization and endocytosis. Traffic 20, 121–129 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Heuninck, J. et al. Context-dependent signaling of CXC chemokine receptor 4 and atypical chemokine receptor 3. Mol. Pharmacol. 96, 778–793 (2019).

    Article  CAS  PubMed  Google Scholar 

  125. Hauser, A. S. et al. Pharmacogenomics of GPCR drug targets. Cell 172, 41–54.e19 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Retamal, J. S., Ramirez-Garcia, P. D., Shenoy, P. A., Poole, D. P. & Veldhuis, N. A. Internalized GPCRs as potential therapeutic targets for the management of pain. Front. Mol. Neurosci. 12, 273 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Jimenez-Vargas, N. N. et al. Protease-activated receptor-2 in endosomes signals persistent pain of irritable bowel syndrome. Proc. Natl Acad. Sci. USA 115, E7438–E7447 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Jimenez-Vargas, N. N. et al. Endosomal signaling of delta opioid receptors is an endogenous mechanism and therapeutic target for relief from inflammatory pain. Proc. Natl Acad. Sci. USA 117, 15281–15292 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Ramirez-Garcia, P. D. et al. A pH-responsive nanoparticle targets the neurokinin 1 receptor in endosomes to prevent chronic pain. Nat. Nanotechnol. 14, 1150–1159 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Arakaki, A. K. S., Pan, W. A. & Trejo, J. GPCRs in cancer: protease-activated receptors, endocytic adaptors and signaling. Int. J. Mol. Sci. 19, 1886 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  131. Dorsam, R. T. & Gutkind, J. S. G-protein-coupled receptors and cancer. Nat. Rev. Cancer 7, 79–94 (2007).

    Article  CAS  PubMed  Google Scholar 

  132. Gad, A. A. & Balenga, N. The emerging role of adhesion GPCRs in cancer. ACS Pharmacol. Transl. Sci. 3, 29–42 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Usman, S., Khawer, M., Rafique, S., Naz, Z. & Saleem, K. The current status of anti-GPCR drugs against different cancers. J. Pharm. Anal. 10, 517–521 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  134. Lavoie, H., Gagnon, J. & Therrien, M. ERK signalling: a master regulator of cell behaviour, life and fate. Nat. Rev. Mol. Cell Biol. 21, 607–632 (2020).

    Article  CAS  PubMed  Google Scholar 

  135. Schiefermeier, N., Teis, D. & Huber, L. A. Endosomal signaling and cell migration. Curr. Opin. Cell Biol. 23, 615–620 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Matsubayashi, Y., Ebisuya, M., Honjoh, S. & Nishida, E. ERK activation propagates in epithelial cell sheets and regulates their migration during wound healing. Curr. Biol. 14, 731–735 (2004).

    Article  CAS  PubMed  Google Scholar 

  137. Aoki, K. et al. Propagating wave of ERK activation orients collective cell migration. Dev. Cell 43, 305–317.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  138. Malinverno, C. et al. Endocytic reawakening of motility in jammed epithelia. Nat. Mater. 16, 587–596 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Villasenor, R., Nonaka, H., Del Conte-Zerial, P., Kalaidzidis, Y. & Zerial, M. Regulation of EGFR signal transduction by analogue-to-digital conversion in endosomes. eLife 4, e06156 (2015).

    Article  PubMed Central  Google Scholar 

  140. Cullen, P. J. & Steinberg, F. To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat. Rev. Mol. Cell Biol. 19, 679–696 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  142. Tebar, F., Enrich, C., Rentero, C. & Grewal, T. GTPases Rac1 and Ras signaling from endosomes. Prog. Mol. Subcell. Biol. 57, 65–105 (2018).

    Article  CAS  PubMed  Google Scholar 

  143. Horiuchi, H. et al. A novel Rab5 GDP/GTP exchange factor complexed to Rabaptin-5 links nucleotide exchange to effector recruitment and function. Cell 90, 1149–1159 (1997).

    Article  CAS  PubMed  Google Scholar 

  144. Christoforidis, S. et al. Phosphatidylinositol-3-OH kinases are Rab5 effectors. Nat. Cell Biol. 1, 249–252 (1999).

    Article  CAS  PubMed  Google Scholar 

  145. Christoforidis, S., McBride, H. M., Burgoyne, R. D. & Zerial, M. The Rab5 effector EEA1 is a core component of endosome docking. Nature 397, 621–625 (1999).

    Article  CAS  PubMed  Google Scholar 

  146. Cezanne, A., Lauer, J., Solomatina, A., Sbalzarini, I. F. & Zerial, M. A non-linear system patterns Rab5 GTPase on the membrane. eLife 9, e54434 (2020). Using an in vitro reconstituted system with lipid bilayers, this study shows that positive feedback regulatory loops control RAB5 recruitment and activation on early endosomes, determining its patterning on endosomal membranes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Murray, J. T., Panaretou, C., Stenmark, H., Miaczynska, M. & Backer, J. M. Role of Rab5 in the recruitment of hVps34/p150 to the early endosome. Traffic 3, 416–427 (2002).

    Article  CAS  PubMed  Google Scholar 

  148. Edler, E. & Stein, M. Probing the druggability of membrane-bound Rab5 by molecular dynamics simulations. J. Enzyme Inhib. Med. Chem. 32, 434–443 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Munzberg, E. & Stein, M. Structure and dynamics of mono- vs. doubly lipidated Rab5 in membranes. Int. J. Mol. Sci. 20, 4773 (2019).

    Article  PubMed Central  CAS  Google Scholar 

  150. Bucci, C. et al. Co-operative regulation of endocytosis by three Rab5 isoforms. FEBS Lett. 366, 65–71 (1995).

    Article  CAS  PubMed  Google Scholar 

  151. Wainszelbaum, M. J., Proctor, B. M., Pontow, S. E., Stahl, P. D. & Barbieri, M. A. IL4/PGE2 induction of an enlarged early endosomal compartment in mouse macrophages is Rab5-dependent. Exp. Cell Res. 312, 2238–2251 (2006).

    Article  CAS  PubMed  Google Scholar 

  152. Chen, P. I., Kong, C., Su, X. & Stahl, P. D. Rab5 isoforms differentially regulate the trafficking and degradation of epidermal growth factor receptors. J. Biol. Chem. 284, 30328–30338 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Bhattacharya, M. et al. IL-6 and IL-12 specifically regulate the expression of Rab5 and Rab7 via distinct signaling pathways. EMBO J. 25, 2878–2888 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Goryachev, A. B. & Pokhilko, A. V. Dynamics of Cdc42 network embodies a Turing-type mechanism of yeast cell polarity. FEBS Lett. 582, 1437–1443 (2008).

    Article  CAS  PubMed  Google Scholar 

  155. Witte, K., Strickland, D. & Glotzer, M. Cell cycle entry triggers a switch between two modes of Cdc42 activation during yeast polarization. eLife 6, e26722 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Zhou, Y. et al. Lipid-sorting specificity encoded in K-ras membrane anchor regulates signal output. Cell 168, 239–251.e16 (2017). Demonstration that clustering of K-RAS at the plasma membrane leads to the assembly of specific phospholipids into nanoclusters determining K-RAS signalling output.

    Article  CAS  PubMed  Google Scholar 

  157. Halatek, J., Brauns, F. & Frey, E. Self-organization principles of intracellular pattern formation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 373, 20170107 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Goryachev, A. B. & Leda, M. Autoactivation of small GTPases by the GEF-effector positive feedback modules. F1000Res https://doi.org/10.12688/f1000research.20003.1 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  159. Mellman, I. & Nelson, W. J. Coordinated protein sorting, targeting and distribution in polarized cells. Nat. Rev. Mol. Cell Biol. 9, 833–845 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Datta, A., Bryant, D. M. & Mostov, K. E. Molecular regulation of lumen morphogenesis. Curr. Biol. 21, R126–R136 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Jewett, C. E. & Prekeris, R. Insane in the apical membrane: trafficking events mediating apicobasal epithelial polarity during tube morphogenesis. Traffic https://doi.org/10.1111/tra.12579 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  162. Overeem, A. W., Bryant, D. M. & van, I. S. C. Mechanisms of apical-basal axis orientation and epithelial lumen positioning. Trends Cell Biol. 25, 476–485 (2015).

    Article  CAS  PubMed  Google Scholar 

  163. Gandalovicova, A., Vomastek, T., Rosel, D. & Brabek, J. Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget 7, 25022–25049 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Roman-Fernandez, A. & Bryant, D. M. Complex polarity: building multicellular tissues through apical membrane traffic. Traffic 17, 1244–1261 (2016). This study shows that the unusual phospholipid PI(3,4)P2, together with PI(4,5)P2, is found apically enriched during the early phases of lumen formation and controls polarity establishment.

    Article  CAS  PubMed  Google Scholar 

  165. West, J. J. & Harris, T. J. Cadherin trafficking for tissue morphogenesis: control and consequences. Traffic 17, 1233–1243 (2016).

    Article  CAS  PubMed  Google Scholar 

  166. Mrozowska, P. S. & Fukuda, M. Regulation of podocalyxin trafficking by Rab small GTPases in 2D and 3D epithelial cell cultures. J. Cell Biol. 213, 355–369 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Diaz-Diaz, C., Baonza, G. & Martin-Belmonte, F. The vertebrate epithelial apical junctional complex: Dynamic interplay between Rho GTPase activity and cell polarization processes. Biochim. Biophys. Acta Biomembr. 1862, 183398 (2020).

    Article  CAS  PubMed  Google Scholar 

  168. Bryant, D. M. & Mostov, K. E. From cells to organs: building polarized tissue. Nat. Rev. Mol. Cell Biol. 9, 887–901 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Nelson, W. J. Adaptation of core mechanisms to generate cell polarity. Nature 422, 766–774 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Schluter, M. A. & Margolis, B. Apicobasal polarity in the kidney. Exp. Cell Res. 318, 1033–1039 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  171. Bryant, D. M. et al. A molecular network for de novo generation of the apical surface and lumen. Nat. Cell Biol. 12, 1035–1045 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Mangan, A. J. et al. Cingulin and actin mediate midbody-dependent apical lumen formation during polarization of epithelial cells. Nat. Commun. 7, 12426 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Su, T. et al. A kinase cascade leading to Rab11-FIP5 controls transcytosis of the polymeric immunoglobulin receptor. Nat. Cell Biol. 12, 1143–1153 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Klinkert, K., Rocancourt, M., Houdusse, A. & Echard, A. Rab35 GTPase couples cell division with initiation of epithelial apico-basal polarity and lumen opening. Nat. Commun. 7, 11166 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Kinoshita, R., Homma, Y. & Fukuda, M. Rab35-GEFs, DENND1A and folliculin differentially regulate podocalyxin trafficking in two- and three-dimensional epithelial cell cultures. J. Biol. Chem. (2020).

  176. Willenborg, C. et al. Interaction between FIP5 and SNX18 regulates epithelial lumen formation. J. Cell Biol. 195, 71–86 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Roman-Fernandez, A. et al. The phospholipid PI(3,4)P2 is an apical identity determinant. Nat. Commun. 9, 5041 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Martin-Belmonte, F. et al. PTEN-mediated apical segregation of phosphoinositides controls epithelial morphogenesis through Cdc42. Cell 128, 383–397 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Bisi, S. et al. IRSp53 controls plasma membrane shape and polarized transport at the nascent lumen in epithelial tubules. Nat. Commun. 11, 3516 (2020). The I-BAR containing protein, IRSp53, is shown to be an early apical determinant that binds to RAB35 and facilitates the transport and the anchoring of podocalyxin to the apical membrane initiation site, where it also controls the integrity and the shape of the plasma membrane.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Zhang, Y. et al. Biomimetic niches reveal the minimal cues to trigger apical lumen formation in single hepatocytes. Nat. Mater. https://doi.org/10.1038/s41563-020-0662-3 (2020). An elegant study that shows how the density and localization of cadherins, along the initial cell–cell contact, represent the minimal molecular and physical cues to trigger the development of asymmetric lateral hemilumen in hepatocytes.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Li, Q. et al. Extracellular matrix scaffolding guides lumen elongation by inducing anisotropic intercellular mechanical tension. Nat. Cell Biol. 18, 311–318 (2016).

    Article  CAS  PubMed  Google Scholar 

  182. Ferrari, A., Veligodskiy, A., Berge, U., Lucas, M. S. & Kroschewski, R. ROCK-mediated contractility, tight junctions and channels contribute to the conversion of a preapical patch into apical surface during isochoric lumen initiation. J. Cell Sci. 121, 3649–3663 (2008).

    Article  CAS  PubMed  Google Scholar 

  183. Saito, Y., Desai, R. R. & Muthuswamy, S. K. Reinterpreting polarity and cancer: The changing landscape from tumor suppression to tumor promotion. Biochim. Biophys. Acta Rev. Cancer 1869, 103–116 (2018).

    Article  CAS  PubMed  Google Scholar 

  184. Vladar, E. K., Antic, D. & Axelrod, J. D. Planar cell polarity signaling: the developing cell’s compass. Cold Spring Harb. Perspect. Biol. 1, a002964 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  185. Simons, M. & Mlodzik, M. Planar cell polarity signaling: from fly development to human disease. Annu. Rev. Genet. 42, 517–540 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Strutt, H. & Strutt, D. Asymmetric localisation of planar polarity proteins: Mechanisms and consequences. Semin. Cell Dev. Biol. 20, 957–963 (2009).

    Article  CAS  PubMed  Google Scholar 

  187. Maung, S. M. & Jenny, A. Planar cell polarity in Drosophila. Organogenesis 7, 165–179 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  188. Xie, Y., Miao, H. & Blankenship, J. T. Membrane trafficking in morphogenesis and planar polarity. Traffic https://doi.org/10.1111/tra.12580 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  189. Butler, M. T. & Wallingford, J. B. Planar cell polarity in development and disease. Nat. Rev. Mol. Cell Biol. 18, 375–388 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Voiculescu, O., Bertocchini, F., Wolpert, L., Keller, R. E. & Stern, C. D. The amniote primitive streak is defined by epithelial cell intercalation before gastrulation. Nature 449, 1049–1052 (2007).

    Article  CAS  PubMed  Google Scholar 

  191. Takeichi, M. Dynamic contacts: rearranging adherens junctions to drive epithelial remodelling. Nat. Rev. Mol. Cell Biol. 15, 397–410 (2014).

    Article  CAS  PubMed  Google Scholar 

  192. Harris, T. J. C. Sculpting epithelia with planar polarized actomyosin networks: Principles from Drosophila. Semin. Cell Dev. Biol. 81, 54–61 (2018).

    Article  CAS  PubMed  Google Scholar 

  193. Pare, A. C. & Zallen, J. A. Cellular, molecular, and biophysical control of epithelial cell intercalation. Curr. Top. Dev. Biol. 136, 167–193 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Truong Quang, B. A., Mani, M., Markova, O., Lecuit, T. & Lenne, P. F. Principles of E-cadherin supramolecular organization in vivo. Curr. Biol. 23, 2197–2207 (2013).

    Article  CAS  PubMed  Google Scholar 

  195. Levayer, R., Pelissier-Monier, A. & Lecuit, T. Spatial regulation of Dia and Myosin-II by RhoGEF2 controls initiation of E-cadherin endocytosis during epithelial morphogenesis. Nat. Cell Biol. 13, 529–540 (2011).

    Article  CAS  PubMed  Google Scholar 

  196. Pope, K. L. & Harris, T. J. Control of cell flattening and junctional remodeling during squamous epithelial morphogenesis in Drosophila. Development 135, 2227–2238 (2008).

    Article  CAS  PubMed  Google Scholar 

  197. Cavanaugh, K. E., Staddon, M. F., Munro, E., Banerjee, S. & Gardel, M. L. RhoA mediates epithelial cell shape changes via mechanosensitive endocytosis. Dev. Cell 52, 152–166.e5 (2020). This study (together with work by Sumi et al.198) illustrates how the pulsating activity of RHOA-mediated contractility, in model epithelial tissues, leads to fluctuation in junctional length and that the shortening of the junction requires formin-mediated E-cadherin clustering and dynamin-dependent endocytosis.

    Article  CAS  PubMed  Google Scholar 

  198. Sumi, A. et al. Adherens junction length during tissue contraction is controlled by the mechanosensitive activity of actomyosin and junctional recycling. Dev. Cell 47, 453–463.e3 (2018). Shows that, during amnioserosa contraction in D. melanogaster, adherens junctions reduce their length in coordination with the shrinkage of apical cell area, maintaining a nearly constant junctional straightness, which is ensured by the endocytic machinery that removes excess plasma membrane.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Miao, H., Vanderleest, T. E., Jewett, C. E., Loerke, D. & Blankenship, J. T. Cell ratcheting through the Sbf RabGEF directs force balancing and stepped apical constriction. J. Cell Biol. 218, 3845–3860 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Jewett, C. E. et al. Planar polarized Rab35 functions as an oscillatory ratchet during cell intercalation in the Drosophila epithelium. Nat. Commun. 8, 476 (2017). By examining RAB protein distributions during cell intercalation in D. melanogaster epithelial tissue remodelling, RAB35-mediated endocytosis of plasma membrane at junctions is found to serve as a unique ratcheting device that directs progressive interface contraction.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Kouranti, I., Sachse, M., Arouche, N., Goud, B. & Echard, A. Rab35 regulates an endocytic recycling pathway essential for the terminal steps of cytokinesis. Curr. Biol. 16, 1719–1725 (2006).

    Article  CAS  PubMed  Google Scholar 

  202. Corallino, S. et al. A RAB35-p85/PI3K axis controls oscillatory apical protrusions required for efficient chemotactic migration. Nat. Commun. 9, 1475 (2018). Demonstration that RAB35 plays a critical role in regulating the formation of oscillatory, apical circular ruffles that act as steering devices during chemotaxis and promote efficient migration and invasion in breast cancer.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Wheeler, D. B., Zoncu, R., Root, D. E., Sabatini, D. M. & Sawyers, C. L. Identification of an oncogenic RAB protein. Science 350, 211–217 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Shaughnessy, R. & Echard, A. Rab35 GTPase and cancer: Linking membrane trafficking to tumorigenesis. Traffic 19, 247–252 (2018).

    Article  CAS  PubMed  Google Scholar 

  205. Rainero, E. Extracellular matrix internalization links nutrient signalling to invasive migration. Int. J. Exp. Pathol. 99, 4–9 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Yong, C. Q. Y. & Tang, B. L. Cancer-driving mutations and variants of components of the membrane trafficking core machinery. Life Sci. 264, 118662 (2021).

    Article  CAS  PubMed  Google Scholar 

  207. Bendris, N. & Schmid, S. L. Endocytosis, metastasis and beyond: multiple facets of SNX9. Trends Cell Biol. 27, 189–200 (2017).

    Article  CAS  PubMed  Google Scholar 

  208. Bisi, S. et al. Membrane and actin dynamics interplay at lamellipodia leading edge. Curr. Opin. Cell Biol. 25, 565–573 (2013).

    Article  CAS  PubMed  Google Scholar 

  209. Caswell, P. T., Vadrevu, S. & Norman, J. C. Integrins: masters and slaves of endocytic transport. Nat. Rev. Mol. Cell Biol. 10, 843–853 (2009).

    Article  CAS  PubMed  Google Scholar 

  210. De Franceschi, N., Hamidi, H., Alanko, J., Sahgal, P. & Ivaska, J. Integrin traffic - the update. J. Cell Sci. 128, 839–852 (2015).

    PubMed  PubMed Central  Google Scholar 

  211. Montell, D. J., Yoon, W. H. & Starz-Gaiano, M. Group choreography: mechanisms orchestrating the collective movement of border cells. Nat. Rev. Mol. Cell Biol. 13, 631–645 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Cheung, K. J. et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc. Natl Acad. Sci. USA 113, E854–E863 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Cheung, K. J., Gabrielson, E., Werb, Z. & Ewald, A. J. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell 155, 1639–1651 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Hakim, V. & Silberzan, P. Collective cell migration: a physics perspective. Rep. Prog. Phys. 80, 076601 (2017).

    Article  PubMed  CAS  Google Scholar 

  215. Tambe, D. T. et al. Collective cell guidance by cooperative intercellular forces. Nat. Mater. 10, 469–475 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Peglion, F., Llense, F. & Etienne-Manneville, S. Adherens junction treadmilling during collective migration. Nat. Cell Biol. 16, 639–651 (2014).

    Article  CAS  PubMed  Google Scholar 

  217. Theveneau, E. & Mayor, R. Neural crest delamination and migration: from epithelium-to-mesenchyme transition to collective cell migration. Dev. Biol. 366, 34–54 (2012).

    Article  CAS  PubMed  Google Scholar 

  218. Lin, M. E., Herr, D. R. & Chun, J. Lysophosphatidic acid (LPA) receptors: signaling properties and disease relevance. Prostaglandins Other Lipid Mediat. 91, 130–138 (2010).

    Article  CAS  PubMed  Google Scholar 

  219. Kuriyama, S. et al. In vivo collective cell migration requires an LPAR2-dependent increase in tissue fluidity. J. Cell Biol. 206, 113–127 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Muinonen-Martin, A. J. et al. Melanoma cells break down LPA to establish local gradients that drive chemotactic dispersal. PLoS Biol. 12, e1001966 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  221. Juin, A. et al. N-WASP control of LPAR1 trafficking establishes response to self-generated LPA gradients to promote pancreatic cancer cell metastasis. Dev. Cell 51, 431–445.e7 (2019). Demonstration that the promoter of actin nucleation, N-WASP (encoded by WASL), drives the trafficking of LPA receptors to control cellular responses to self-generated gradients and to enhance metastatic spreading of pancreatic ductal carcinomas.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Leyton-Puig, D. et al. Flat clathrin lattices are dynamic actin-controlled hubs for clathrin-mediated endocytosis and signalling of specific receptors. Nat. Commun. 8, 16068 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Park, J. A. et al. Unjamming and cell shape in the asthmatic airway epithelium. Nat. Mater. 14, 1040–1048 (2015). A seminal paper showing the physical principles governing the transition from a solid, jammed to a collectively moving and unjammed pseudostratified human bronchial epithelial system from asthmatic patients.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Sadati, M., Nourhani, A., Fredberg, J. J. & Taheri Qazvini, N. Glass-like dynamics in the cell and in cellular collectives. Wiley Interdiscip. Rev. Syst. Biol. Med. 6, 137–149 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Sadati, M., Taheri Qazvini, N., Krishnan, R., Park, C. Y. & Fredberg, J. J. Collective migration and cell jamming. Differentiation 86, 121–125 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Park, J. A., Atia, L., Mitchel, J. A., Fredberg, J. J. & Butler, J. P. Collective migration and cell jamming in asthma, cancer and development. J. Cell Sci. 129, 3375–3383 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Atia, L. et al. Geometric constraints during epithelial jamming. Nat. Phys. 14, 613–620 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Bi, D., Yang, X., Marchetti, M. C. & Manning, M. L. Motility-driven glass and jamming transitions in biological tissues. Phys. Rev. X 6, 021011 (2016).

    PubMed  PubMed Central  Google Scholar 

  229. Garcia, S. et al. Physics of active jamming during collective cellular motion in a monolayer. Proc. Natl Acad. Sci. USA 112, 15314–15319 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Palamidessi, A. et al. Unjamming overcomes kinetic and proliferation arrest in terminally differentiated cells and promotes collective motility of carcinoma. Nat. Mater. 18, 1252–1263 (2019).

    Article  CAS  PubMed  Google Scholar 

  231. Ilina, O. et al. Cell-cell adhesion and 3D matrix confinement determine jamming transitions in breast cancer invasion. Nat. Cell Biol. 22, 1103–1115 (2020). In this work, the authors show that cell crowding induced by matrix confinement and cell–cell adhesion modulate the jamming transition during breast cancer invasion.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Mongera, A. et al. A fluid-to-solid jamming transition underlies vertebrate body axis elongation. Nature 561, 401–405 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Mitchel, J. A. et al. In primary airway epithelial cells, the unjamming transition is distinct from the epithelial-to-mesenchymal transition. Nat. Commun. 11, 5053 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Greenburg, G. & Hay, E. D. Epithelia suspended in collagen gels can lose polarity and express characteristics of migrating mesenchymal cells. J. Cell Biol. 95, 333–339 (1982).

    Article  CAS  PubMed  Google Scholar 

  235. Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. Emt: 2016. Cell 166, 21–45 (2016).

    Article  CAS  PubMed  Google Scholar 

  236. Aiello, N. M. & Kang, Y. Context-dependent EMT programs in cancer metastasis. J. Exp. Med. 216, 1016–1026 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Bakir, B., Chiarella, A. M., Pitarresi, J. R. & Rustgi, A. K. EMT, MET, plasticity, and tumor metastasis. Trends Cell Biol. 30, 764–776 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  238. Pastushenko, I. & Blanpain, C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 29, 212–226 (2019).

    Article  CAS  PubMed  Google Scholar 

  239. Dongre, A. & Weinberg, R. A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 20, 69–84 (2019).

    Article  CAS  PubMed  Google Scholar 

  240. Mani, S. A. et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. van Staalduinen, J., Baker, D., Ten Dijke, P. & van Dam, H. Epithelial-mesenchymal-transition-inducing transcription factors: new targets for tackling chemoresistance in cancer? Oncogene 37, 6195–6211 (2018).

    Article  PubMed  CAS  Google Scholar 

  242. Shibue, T. & Weinberg, R. A. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 14, 611–629 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  243. Corallino, S., Malabarba, M. G., Zobel, M., Di Fiore, P. P. & Scita, G. Epithelial-to-mesenchymal plasticity harnesses endocytic circuitries. Front. Oncol. 5, 45 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  244. Yang, J. et al. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 21, 341–352 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  245. Stemmler, M. P., Eccles, R. L., Brabletz, S. & Brabletz, T. Non-redundant functions of EMT transcription factors. Nat. Cell Biol. 21, 102–112 (2019).

    Article  CAS  PubMed  Google Scholar 

  246. Skrypek, N., Goossens, S., De Smedt, E., Vandamme, N. & Berx, G. Epithelial-to-mesenchymal transition: epigenetic reprogramming driving cellular plasticity. Trends Genet. 33, 943–959 (2017).

    Article  CAS  PubMed  Google Scholar 

  247. Lamouille, S., Xu, J. & Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 15, 178–196 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Pastushenko, I. et al. Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018). The first in vivo demonstration that, in tumours, subpopulations that exhibit all different EMT stages, from epithelial to mesenchymal, can be identified through intermediate different hybrid states.

    Article  CAS  PubMed  Google Scholar 

  249. Aiello, N. M. et al. EMT subtype influences epithelial plasticity and mode of cell migration. Dev. Cell 45, 681–695.e4 (2018). Through an in vivo approach, in a mouse model of pancreatic ductal adenocarcinoma, it is shown that EMT can proceed through endocytosis rather than transcriptional reprogramming, leading to a partial EMT phenotype.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Reichert, M. et al. Regulation of epithelial plasticity determines metastatic organotropism in pancreatic cancer. Dev. Cell 45, 696–711.e8 (2018). By using several mouse models of pancreatic ductal adenocarcinoma, the authors show that the organotropic metastatic preference is a function of the degree of EMT (full blown EMT versus partial EMT).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Vieira, A. V., Lamaze, C. & Schmid, S. L. Control of EGF receptor signaling by clathrin-mediated endocytosis. Science 274, 2086–2089 (1996).

    Article  CAS  PubMed  Google Scholar 

  252. Khan, M. N., Savoie, S., Bergeron, J. J. & Posner, B. I. Characterization of rat liver endosomal fractions. In vivo activation of insulin-stimulable receptor kinase in these structures. J. Biol. Chem. 261, 8462–8472 (1986).

    Article  CAS  PubMed  Google Scholar 

  253. Lai, W. H., Cameron, P. H., Doherty, J. J. 2nd, Posner, B. I. & Bergeron, J. J. Ligand-mediated autophosphorylation activity of the epidermal growth factor receptor during internalization. J. Cell Biol. 109, 2751–2760 (1989).

    Article  CAS  PubMed  Google Scholar 

  254. Scita, G. & Di Fiore, P. P. The endocytic matrix. Nature 463, 464–473 (2010).

    Article  CAS  PubMed  Google Scholar 

  255. Clevers, H. Modeling development and disease with organoids. Cell 165, 1586–1597 (2016).

    Article  CAS  PubMed  Google Scholar 

  256. Mayle, K. M., Le, A. M. & Kamei, D. T. The intracellular trafficking pathway of transferrin. Biochim. Biophys. Acta 1820, 264–281 (2012).

    Article  CAS  PubMed  Google Scholar 

  257. Maurer, M. E. & Cooper, J. A. The adaptor protein Dab2 sorts LDL receptors into coated pits independently of AP-2 and ARH. J. Cell Sci. 119, 4235–4246 (2006).

    Article  CAS  PubMed  Google Scholar 

  258. Mishra, S. K. et al. Disabled-2 exhibits the properties of a cargo-selective endocytic clathrin adaptor. EMBO J. 21, 4915–4926 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Mishra, S. K., Watkins, S. C. & Traub, L. M. The autosomal recessive hypercholesterolemia (ARH) protein interfaces directly with the clathrin-coat machinery. Proc. Natl Acad. Sci. USA 99, 16099–16104 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Morris, S. M. & Cooper, J. A. Disabled-2 colocalizes with the LDLR in clathrin-coated pits and interacts with AP-2. Traffic 2, 111–123 (2001).

    Article  CAS  PubMed  Google Scholar 

  261. Tao, W., Moore, R., Meng, Y., Smith, E. R. & Xu, X. X. Endocytic adaptors Arh and Dab2 control homeostasis of circulatory cholesterol. J. Lipid Res. 57, 809–817 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. He, G. et al. ARH is a modular adaptor protein that interacts with the LDL receptor, clathrin, and AP-2. J. Biol. Chem. 277, 44044–44049 (2002).

    Article  CAS  PubMed  Google Scholar 

  263. Beglova, N. & Blacklow, S. C. The LDL receptor: how acid pulls the trigger. Trends Biochem. Sci. 30, 309–317 (2005).

    Article  CAS  PubMed  Google Scholar 

  264. Renard, H. F. et al. Endophilin-A2 functions in membrane scission in clathrin-independent endocytosis. Nature 517, 493–496 (2015).

    Article  CAS  PubMed  Google Scholar 

  265. Simunovic, M. et al. Friction mediates scission of tubular membranes scaffolded by BAR proteins. Cell 170, 172–184.e11 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Galperin, E. & Sorkin, A. Endosomal targeting of MEK2 requires RAF, MEK kinase activity and clathrin-dependent endocytosis. Traffic 9, 1776–1790 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Pinilla-Macua, I., Watkins, S. C. & Sorkin, A. Endocytosis separates EGF receptors from endogenous fluorescently labeled HRas and diminishes receptor signaling to MAP kinases in endosomes. Proc. Natl Acad. Sci. USA 113, 2122–2127 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Sigismund, S. et al. Threshold-controlled ubiquitination of the EGFR directs receptor fate. EMBO J. 32, 2140–2157 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Rochman, Y., Spolski, R. & Leonard, W. J. New insights into the regulation of T cells by gamma(c) family cytokines. Nat. Rev. Immunol. 9, 480–490 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Basquin, C. et al. Membrane protrusion powers clathrin-independent endocytosis of interleukin-2 receptor. EMBO J. 34, 2147–2161 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Grassart, A., Dujeancourt, A., Lazarow, P. B., Dautry-Varsat, A. & Sauvonnet, N. Clathrin-independent endocytosis used by the IL-2 receptor is regulated by Rac1, Pak1 and Pak2. EMBO Rep. 9, 356–362 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Sauvonnet, N., Dujeancourt, A. & Dautry-Varsat, A. Cortactin and dynamin are required for the clathrin-independent endocytosis of gammac cytokine receptor. J. Cell Biol. 168, 155–163 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Blasky, A. J., Mangan, A. & Prekeris, R. Polarized protein transport and lumen formation during epithelial tissue morphogenesis. Annu. Rev. Cell Dev. Biol. 31, 575–591 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Winter, J. F. et al. Caenorhabditis elegans screen reveals role of PAR-5 in RAB-11-recycling endosome positioning and apicobasal cell polarity. Nat. Cell Biol. 14, 666–676 (2012).

    Article  CAS  PubMed  Google Scholar 

  275. Apodaca, G., Gallo, L. I. & Bryant, D. M. Role of membrane traffic in the generation of epithelial cell asymmetry. Nat. Cell Biol. 14, 1235–1243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Henry, L. & Sheff, D. R. Rab8 regulates basolateral secretory, but not recycling, traffic at the recycling endosome. Mol. Biol. Cell 19, 2059–2068 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Babbey, C. M. et al. Rab10 regulates membrane transport through early endosomes of polarized Madin-Darby canine kidney cells. Mol. Biol. Cell 17, 3156–3175 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Lock, J. G. & Stow, J. L. Rab11 in recycling endosomes regulates the sorting and basolateral transport of E-cadherin. Mol. Biol. Cell 16, 1744–1755 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Duman, J. G., Tyagarajan, K., Kolsi, M. S., Moore, H. P. & Forte, J. G. Expression of rab11a N124I in gastric parietal cells inhibits stimulatory recruitment of the H+-K+-ATPase. Am. J. Physiol. 277, C361–C372 (1999).

    Article  CAS  PubMed  Google Scholar 

  280. Li, D., Mangan, A., Cicchini, L., Margolis, B. & Prekeris, R. FIP5 phosphorylation during mitosis regulates apical trafficking and lumenogenesis. EMBO Rep. 15, 428–437 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  281. Roland, J. T. et al. Rab GTPase-Myo5B complexes control membrane recycling and epithelial polarization. Proc. Natl Acad. Sci. USA 108, 2789–2794 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Mendoza, M. C. et al. ERK-MAPK drives lamellipodia protrusion by activating the WAVE2 regulatory complex. Mol. Cell 41, 661–671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  283. Mendoza, M. C., Vilela, M., Juarez, J. E., Blenis, J. & Danuser, G. ERK reinforces actin polymerization to power persistent edge protrusion during motility. Sci. Signal. 8, ra47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  284. Farooqui, R. & Fenteany, G. Multiple rows of cells behind an epithelial wound edge extend cryptic lamellipodia to collectively drive cell-sheet movement. J. Cell Sci. 118, 51–63 (2005).

    Article  CAS  PubMed  Google Scholar 

  285. Giavazzi, F. et al. Flocking transitions in confluent tissues. Soft Matter 14, 3471–3477 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Giavazzi, F. et al. Giant fluctuations and structural effects in a flocking epithelium. J. Phys. D Appl. Phys. 50, 384003 (2017).

    Article  CAS  Google Scholar 

  287. Boucrot, E. & Kirchhausen, T. Endosomal recycling controls plasma membrane area during mitosis. Proc. Natl Acad. Sci. USA 104, 7939–7944 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Tacheva-Grigorova, S. K., Santos, A. J., Boucrot, E. & Kirchhausen, T. Clathrin-mediated endocytosis persists during unperturbed mitosis. Cell Rep. 4, 659–668 (2013).

    Article  CAS  PubMed  Google Scholar 

  289. Aguet, F. et al. Membrane dynamics of dividing cells imaged by lattice light-sheet microscopy. Mol. Biol. Cell 27, 3418–3435 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Dix, C. L. et al. The role of mitotic cell-substrate adhesion re-modeling in animal cell division. Dev. Cell 45, 132–145.e3 (2018).

    Article  CAS  PubMed  Google Scholar 

  291. Jones, M. C., Askari, J. A., Humphries, J. D. & Humphries, M. J. Cell adhesion is regulated by CDK1 during the cell cycle. J. Cell Biol. 217, 3203–3218 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Lock, J. G. et al. Reticular adhesions are a distinct class of cell-matrix adhesions that mediate attachment during mitosis. Nat. Cell Biol. 20, 1290–1302 (2018). Mitotic matrix adhesion sites, termed ‘reticular adhesions’, are characterized in this study, showing that they are morphologically, dynamically and molecularly distinct from classical focal adhesions, being enriched in components of the clathrin machinery.

    Article  CAS  PubMed  Google Scholar 

  293. Zaidel-Bar, R. Atypical matrix adhesions guide cell division. Nat. Cell Biol. 20, 1233–1235 (2018).

    Article  CAS  PubMed  Google Scholar 

  294. Elkhatib, N. et al. Tubular clathrin/AP-2 lattices pinch collagen fibers to support 3D cell migration. Science 356, eaal4713 (2017).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Maria Grazia Malabarba for support in figure design and Rosalind Gunby for critically editing the manuscript before submission. Work in the authors’ labs is supported by: Associazione Italiana per la Ricerca sul Cancro (AIRC IG 24415 to SS, AIRC IG 22811 to LL, AIRC IG 18621 and 5XMille 22759 to GS, and AIRC IG 18988, AIRC IG 23060 and MCO 10000 to PPDF); the Worldwide Cancer Research (20-0094 to SS), the Italian Ministry of University and Scientific Research (PRIN 2017, Prot. 2017E5L5P3 to SS; Prot. 2017HWTP2K to GS; PRIN 2015 Prot. 2015XS92CC to PPDF); the University of Milan (PSR2019 to SS); the FPRC 5×1000 Ministero Salute 2017 (to LL); the Italian Ministry of Health (RF-2016-02361540 to PPDF).

We apologize to the many colleagues whose excellent work could not be reviewed or cited for lack of space. An additional number of primary research papers and reviews is included in the Supplementary Information.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Pier Paolo Di Fiore.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks V. Haucke and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Receptor tyrosine kinases

(RTKs). A family of plasma membrane proteins (~60 genes in humans) that function as high affinity binding sites for growth factors and cytokines and transduce signals intracellularly through their intrinsic tyrosine kinase activity.

G protein-coupled receptors

(GPCRs). A vast family of plasma membrane receptors (more than 800 genes in humans) characterized by seven transmembrane regions. They transduce signals through a variety of modes, among which the best characterized is the coupling with heterotrimeric G proteins.

Arrestins

A family of proteins that act as multifunctional scaffolding proteins, regulating the trafficking and signalling of transmembrane receptors, particularly of GPCRs. They are involved in receptor desensitization, endocytosis and ubiquitylation. They can also function as positive effectors of GPCRs through their scaffolding abilities. The arrestin family comprises visual arrestins, β-arrestins (non-visual arrestins) and α-arrestins.

AKT kinase

The three members of the human AKT serine-threonine protein kinase family are often referred to as protein kinase Bα (PKBα), PKBβ and PKBγ. These proteins are phosphorylated by phosphoinositide 3-kinase (PI3K). AKT–PI3K forms a key component of many signalling pathways that involves the binding of membrane-bound ligands such as RTKs.

Epsin family of endocytic adaptor proteins

A family of endocytic proteins composed of three paralogs: EPN1, EPN2 and EPN3, characterized by the presence of an epsin N-terminal homology domain involved in phosphoinositide binding at the plasma membrane, ubiquitin binding motifs and motifs that bind to clathrin, AP2 and other endocytic proteins. They are involved in both clathrin-mediated endocytosis, where they play a role in clathrin-coat assembly and cargo recruitment, and in the non-clathrin endocytosis of epidermal growth factor receptor.

Epithelial–mesenchymal transition

(EMT). A process, of great relevance in embryogenesis, through which epithelial cells lose polarity and cell–cell adhesion contacts (sessile state) to acquire characteristics of migratory mesenchymal-like cells. In physiology, typically, after migrating, these cells re-acquire an epithelial phenotype through the opposite process of mesenchymal–epithelial transition.

Death receptors

Type I transmembrane proteins belonging to the tumour necrosis factor/nerve growth factor superfamily. They are activated upon binding to various agonists (such as FASLG, TNFA or TRAIL). They typically trigger the so-called apoptotic extrinsic pathway, yet they can also activate multiple alternative signalling pathways with opposing outcomes (survival/proliferation versus cell death) depending on the cell context.

Caveolae

Small flask-shaped invaginations of the plasma membrane (50–80 nm) that can be morphologically identified by the presence of coat-like proteins (caveolins) and that are particularly abundant in tissues involved in lipid homeostasis or subjected to mechanical challenges like adipocytes, muscle cells and endothelial cells.

Vinculin

A protein involved in the formation of focal adhesions that links surface structures (integrins) to the actin cytoskeleton (through binding to F-actin).

Focal adhesions

Cell-to-matrix adhesion structures involved in the transmission and regulation of signals between the extracellular matrix and the intracellular environment. They are large and dynamic protein complexes established through integrins (which bind to the extracellular matrix), vinculin, F-actin and several regulatory components (up to 100 different proteins, according to the state of the cell). Focal adhesions have roles in signal transduction, cell motility, cell cycle regulation and several other cellular phenotypes. They represent one of the main sensors/effectors in cellular mechanosensing.

Galectins

A class of proteins that bind specifically to β-galactoside sugars such as N-acetyl-lactosamine. Galectins are secreted in the extracellular space, where they encounter galactose-containing glycoproteins and glycolipids. The binding of galectins to glycosylated proteins, such as CD44 and α5β1 integrin, triggers galectin oligomerization, which allows their interaction with glycosphingolipids and the generation of plasma membrane curvature, leading to the formation of clathrin-independent endocytic carriers.

Tip cells

During angiogenesis, new vessels that sprout from existing ones are guided by a leader cell that drives the extension of the sprout and senses the environment for guidance cues.

ERK signalling

Signalling mediated by the activation of the extracellular signal-regulated kinases (ERKs, also called mitogen-activated protein kinases or MAPKs). This signalling is mediated by the sequential activation of the small GTPase RAS and a cascade of kinases (RAF, MEK and ERK1,2) that transduce a signal from a receptor, located on the cell surface or on endosomes, to regulate a number of fundamental biological functions, including cell proliferation, differentiation and migration.

PAK

A family of serine/threonine protein kinases that includes six members in mammals. They serve as targets for the small GTPases CDC42 and RAC and have been implicated in a wide range of biological activities.

PTEN

A lipid phosphatase (phosphatidylinositol 3,4,5 triphosphate 3-phosphatase). It catalyses the conversion of PI(3,4,5)P3 to PI(4,5)P2, thereby antagonizing the action of PI3K and the activation of AKT. It represents one of the most frequently lost tumour suppressors in human cancers.

AMPK

AMP-activated protein kinase or 5’ adenosine monophosphate-activated protein kinase. It is a heterotrimeric protein complex endowed with serine/threonine kinase activity that regulates the energy metabolism, mostly acting on glucose and fatty acid metabolism.

RAC1

A member of RHO subfamily of small GTPases that plays a central role in controlling the activity of protein complexes that are necessary to remodel the actin cytoskeleton during migration.

p38

A member of a class of MAPKs that are responsive to stress stimuli, such as cytokines, ultraviolet irradiation, heat shock and osmotic shock, and are involved in cell differentiation, apoptosis and autophagy.

JNK

JNK (or c-Jun N-terminal Kinase) is a member of a family of protein kinases, which play a central role in stress signalling pathways implicated in gene expression, neuronal plasticity, regeneration, cell death and regulation of cellular senescence.

RAB GTPases

A subfamily of small GTPases that includes more than 70 members in mammals and regulates several key steps of membrane trafficking, including vesicle formation, vesicle movement along actin and tubulin networks, and membrane fusion.

GEF

This term broadly defines a vast group of proteins (frequently unrelated) that all possess Guanine Nucleotide Exchange Factor activity, that is, the ability to convert G proteins from an inactive GDP-bound to an active GTP-bound form.

Jammed epithelial monolayers

The dynamics of epithelia has been described in terms of jamming transitions. During this transition, collective motion ceases, cells can no longer exchange neighbours, and monolayers become static and rigid, displaying a behaviour similar to that of ensembles of dense and packed inactive particles such as coffee in a chute or sand in a pile.

Midbody

Central region of the thin cytoplasmic bridge that connects cells at the end of cytokinesis. It consists mostly of microtubules, together with various other types of proteins (400–500). It functions as a platform to mediate abscission, the process of severing the intercellular bridge. It is also endowed with numerous other functions, including the determination of cell fate and asymmetric post-abscission signal transduction.

WAVE

A key component of a pentameric actin nucleation promoting complex that acts downstream of the GTPase RAC and is necessary for activating the Arp2/3 complex for the generation of branched actin networks.

Tight junction

A cell-to-cell junction formed by a multiprotein complex. This type of junction is established through homotypic interactions between adhesion molecules (occludins, claudins, JAMs) present on the surface of abutting cells. Tight junctions mark the border between the apical and the basolateral surfaces in epithelial cells and control the formation of functionally distinct apical domains. They are also present in endothelial cells and astrocytes and establish the blood–brain barrier. One of their major function is to seal the epithelia by preventing the leakage of water and small molecular weight solutes.

Arp2/3 complex

A seven-subunit complex that, upon activation, promotes the branched elongation of the actin network by binding to the side of mother filaments.

Crumbs polarity complex

A multiprotein complex composed of three members originally identified in Drosophila melanogaster, Crumbs, Pals1 and PatJ. This complex plays a key role in specifying the apical plasma membrane domain of epithelial cells and in controlling cell shape in both invertebrates and vertebrates.

Transcytosis

A process in which molecules are transported across cellular barriers. It involves the endocytosis of molecules (typically plasma membrane proteins or extracellular molecules captured through interaction with surface receptors) at one side of the cell and their vesicle-mediated transport to another side, where they are released through exocytosis. It contributes to the establishment of apical–basal cell polarity by transferring transmembrane proteins between distinct plasma membrane domains. It is involved in many other processes, for instance, in the crossing of the blood–brain barrier.

Exocyst

An octameric protein complex involved in vesicle trafficking, specifically in the tethering and spatial targeting of vesicles to the plasma membrane prior to vesicle fusion.

Annexin 2

A 36-kDa calcium-dependent, phospholipid-binding protein that functions in promoting the exocytosis of intracellular proteins to the extracellular space.

CDC42 apical polarity complex

CDC42 is a highly conserved RHO-family GTPase that regulates cell polarity in many eukaryotes. It directly interacts with PAR6 and regulates, through this protein, the activity of the atypical protein kinase C (aPKC).

I-BAR domain

BAR domains are banana-shaped protein domains capable of sensing membrane curvature by binding preferentially to positively curved membranes and named after three proteins in which they were originally identified: BIN1 (bridging interactor 1), AMPH (amphiphysin) and Rvs167 (the yeast homolog of amphiphysin). In contrast to the banana-shaped BAR domains, the I-BAR (which stand for inverse BAR) domain is a zeppelin-shaped structure with convex geometry, thus generating negative membrane curvature.

Adherens junctions

Cadherin-based cell-to-cell junctions present in epithelial and endothelial cells, frequently in a more basal position with respect to tight junctions.

Isochoric

A process in which the volume of a closed system does not change. It is synonym of isovolumetric.

Germband

In Drosophila melanogaster, the ventral part of the embryo that forms during gastrulation and gives rise to the segmented trunk of the animal (gnathal, thoracic, abdominal segments). It includes the mesoderm, ventral ectoderm and dorsal epidermis but excludes the dorsal-most tissue of the embryo, the amnioserosa.

Amnioserosa

In Drosophila melanogaster, a short-lived extraembryonic tissue with a critical role in dorsal closure and other early developmental morphogenetic events.

Border cells

A cluster of cells that migrate from the anterior tip of the Drosophila melanogaster egg chamber to the border of the oocyte at stage 9 of oogenesis. These cells perform a stereotypical collective migration on the intervening nurse cells and reach the oocyte. They are required for the formation of the micropyle, the eggshell structure through which sperm enters the egg.

Lamellipodia

Thin membrane protrusion present at the leading edge of migrating cells, mostly constituted by a flat network of actin.

Treadmilling

A process characterizing filamentous multimeric protein structures within the cell and mostly used in reference to filamentous actin (F-actin). When actin subunits (G-actin) are constantly added at one end of the filament and removed from the opposite one, the net effect is the treadmilling of the filament which is used, for instance, to generate motion. The term is also used, more generally, for other biological processes in which the treadmilling of molecules or organelles occurs.

Neural crest

A temporary group of cells established during vertebrate development, which forms after gastrulation at the border between the neural plate and the surrounding ectoderm. After closure of the neural tube (due to the folding of the neural plate into itself), the neural crest runs along the roof plate of the neural tube. At this stage, neural crest cells undergo EMT and migrate to the periphery, where they give origin to various cell lineages.

Ascitic fluid

An abnormal accumulation of fluid in the abdominal cavity frequently caused by liver disease or cirrhosis, cancers (specifically ovarian and colon cancer), and heart failure.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sigismund, S., Lanzetti, L., Scita, G. et al. Endocytosis in the context-dependent regulation of individual and collective cell properties. Nat Rev Mol Cell Biol 22, 625–643 (2021). https://doi.org/10.1038/s41580-021-00375-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-021-00375-5

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