Skip to main navigation menu Skip to main content Skip to site footer

Review article: Biomedical intelligence

Vol. 146 No. 3738 (2016)

When membranes need an ESCRT: endosomal sorting and membrane remodelling in health and disease

  • Victor Alfred
  • Thomas Vaccari
DOI
https://doi.org/10.4414/smw.2016.14347
Cite this as:
Swiss Med Wkly. 2016;146:w14347
Published
11.09.2016

Summary

Originally discovered as regulators of cargo sorting during endosomal trafficking, ESCRT (endosomal sorting complexes required for transport) proteins are emerging as flexible machines that shape the behaviour of membranes throughout the cell. Deregulation of ESCRT activity is observed in neuro-degenerative diseases, virus infection and cancer. However, the mechanisms of pathogenesis involving ESCRTs have not yet fully come into focus. Here, we review the current knowledge of ESCRT function in health and disease and provide educated guesses for future research and focused therapeutic intervention.

References

  1. Cavalier-Smith T. The phagotrophic origin of eukaryotes and phylogenetic classification of Protozoa. Int J Syst Evol Microbiol. 2002;52:297–354.
  2. Dacks JB, Field MC. Evolution of the eukaryotic membrane-trafficking system: origin, tempo and mode. J Cell Sci. 2007;120:2977–85.
  3. Rothman JH, Howald I, Stevens TH. Characterization of genes required for protein sorting and vacuolar function in the yeast Saccharomyces cerevisiae. EMBO J. 1989;8:2057–65.
  4. Bankaitis VA, Johnson LM, Emr SD. Isolation of yeast mutants defective in protein targeting to the vacuole. Proc Natl Acad Sci U S A. 1986;83:9075–9.
  5. Raymond CK, Howald-Stevenson I, Vater CA, Stevens TH. Morphological classification of the yeast vacuolar protein sorting mutants: evidence for a prevacuolar compartment in class E vps mutants. Mol Biol Cell. 1992;3:1389–402.
  6. Katzmann DJ, Babst M, Emr SD. Ubiquitin-Dependent Sorting into the Multivesicular Body Pathway Requires the Function of a Conserved Endosomal Protein Sorting Complex, ESCRT-I. Cell. 2001;106:145–55.
  7. Urbé S. Ubiquitin and endocytic protein sorting. Essays Biochem. 2005;41:81–98.
  8. Raiborg C, Bache KG, Mehlum A, Stang E, Stenmark H. Hrs recruits clathrin to early endosomes. EMBO J. 2001;20:5008–21.
  9. Raiborg C, Bache KG, Gillooly DJ, Madshus IH, Stang E, Stenmark H. Hrs sorts ubiquitinated proteins into clathrin-coated microdomains of early endosomes. Nat Cell Biol. 2002;4:394–8.
  10. Wollert T, Hurley JH. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature. 2010;464:864–9.
  11. Malerød L, Stenmark H. ESCRTing membrane deformation. Cell. 2009;136:15–7.
  12. Teis D, Saksena S, Judson BL, Emr SD. ESCRT-II coordinates the assembly of ESCRT-III filaments for cargo sorting and multivesicular body vesicle formation. EMBO J. 2010;29:871–83.
  13. Shields SB, Oestreich AJ, Winistorfer S, Nguyen D, Payne J a, Katzmann DJ, Piper R. ESCRT ubiquitin-binding domains function cooperatively during MVB cargo sorting. J Cell Biol. 2009;185:213–24.
  14. Mageswaran SK, Dixon MG, Curtiss M, Keener JP, Babst M. Binding to any ESCRT can mediate ubiquitin-independent cargo sorting. Traffic. 2014;15:212–29.
  15. Bache KG, Brech A, Mehlum A, Stenmark H. Hrs regulates multivesicular body formation via ESCRT recruitment to endosomes. J Cell Biol. 2003;162:435–42.
  16. Henne WM, Buchkovich NJ, Zhao Y, Emr SD. The endosomal sorting complex ESCRT-II mediates the assembly and architecture of ESCRT-III helices. Cell. 2012;151:356–71.
  17. Adell MAY, Vogel GF, Pakdel M, Muller M, Lindner H, Hess MW, Teis D. Coordinated binding of Vps4 to ESCRT-III drives membrane neck constriction during MVB vesicle formation. J Cell Biol. 2014;205:33–49.
  18. Yang B, Stjepanovic G, Shen Q, Martin A, Hurley JH. Vps4 disassembles an ESCRT-III filament by global unfolding and processive translocation. Nat Struct Mol Biol. 2015;22:492–8.
  19. Williams RL, Urbé S. The emerging shape of the ESCRT machinery. Nat Rev Mol Cell Biol. 2007;8:355–68.
  20. Hurley JH. The ESCRT complexes. Crit Rev Biochem Mol Biol. 2010;45:463–87.
  21. Wideman JG, Leung KF, Field MC, Dacks JB. The cell biology of the endocytic system from an evolutionary perspective. Cold Spring Harb Perspect Biol. 2014;6:a016998.
  22. Blanc C, Charette SJ, Mattei S, Aubry L, Smith EW, Cosson P, Letourneur F. Dictyostelium Tom1 participates to an ancestral ESCRT-0 complex. Traffic. 2009;10:161–71.
  23. Garrus JE, von Schwedler UK, Pornillos OW, Morham SG, Zavitz KH, Wang HE, et al. Tsg101 and the vacuolar protein sorting pathway are essential for HIV-1 budding. Cell. 2001;107:55–65.
  24. Pornillos O, Alam SL, Davis DR, Sundquist WI. Structure of the Tsg101 UEV domain in complex with the PTAP motif of the HIV-1 p6 protein. Nat Struct Biol. 2002;9:812–7.
  25. Wehman AM, Poggioli C, Schweinsberg P, Grant BD, Nance J. The P4-ATPase TAT-5 inhibits the budding of extracellular vesicles in C. elegans embryos. Curr Biol. 2011;21:1951–9.
  26. Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of TSG101 protein. Proc Natl Acad Sci U S A. 2012;109:4146–51.
  27. Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14:677–85.
  28. Choudhuri K, Llodrá J, Roth EW, Tsai J, Gordo S, Wucherpfennig KW, et al. Polarized release of T-cell-receptor-enriched microvesicles at the immunological synapse. Nature. 2014;507:118–23.
  29. MacDonald C, Payne JA, Aboian M, Smith W, Katzmann DJ, Piper RC. A family of tetraspans organizes cargo for sorting into multivesicular bodies. Dev Cell. 2015;33:328–42.
  30. Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, et al. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126:5553–65.
  31. Kaur J, Debnath J. Autophagy at the crossroads of catabolism and anabolism. Nat Rev Mol Cell Biol. 2015;16:461–72.
  32. Lamb CA, Yoshimori T, Tooze SA. The autophagosome: origins unknown, biogenesis complex. Nat Rev Mol Cell Biol. 2013;14:759–74.
  33. Rusten TE, Vaccari T, Lindmo K, Rodahl LMW, Nezis IP, Sem-Jacobsen C, et al. ESCRTs and Fab1 regulate distinct steps of autophagy. Curr Biol. 2007;17:1817–25.
  34. Roudier N, Lefebvre C, Legouis R. CeVPS-27 is an endosomal protein required for the molting and the endocytic trafficking of the low-density lipoprotein receptor-related protein 1 in Caenorhabditis elegans. Traffic. 2005;6:695–705.
  35. Morelli E, Ginefra P, Mastrodonato V, Beznoussenko G V, Rusten TE, Bilder D, et al. Multiple functions of the SNARE protein Snap29 in autophagy, endocytic, and exocytic trafficking during epithelial formation in Drosophila. Autophagy. 2014;10:2251–68.
  36. Sahu R, Kaushik S, Clement CC, Cannizzo ES, Scharf B, Follenzi A, et al. Microautophagy of cytosolic proteins by late endosomes. Dev Cell. 2011;20:131–9.
  37. Jones CB, Ott EM, Keener JM, Curtiss M, Sandrin V, Babst M. Regulation of membrane protein degradation by starvation-response pathways. Traffic. 2012;13:468–82.
  38. Müller M, Schmidt O, Angelova M, Faserl K, Weys S, Kremser L, et al. The coordinated action of the MVB pathway and autophagy ensures cell survival during starvation. Elife. 2015;4:e07736.
  39. Carlton JG, Martin-Serrano J. Parallels between cytokinesis and retroviral budding: a role for the ESCRT machinery. Science. 2007;316:1908–12.
  40. Lindås A-C, Karlsson EA, Lindgren MT, Ettema TJG, Bernander R. A unique cell division machinery in the Archaea. Proc Natl Acad Sci U S A. 2008;105:18942–6.
  41. Samson RY, Bell SD. Ancient ESCRTs and the evolution of binary fission. Trends Microbiol. 2009;17:507–13.
  42. Vietri M, Schink KO, Campsteijn C, Wegner CS, Schultz SW, Christ L, et al. Spastin and ESCRT-III coordinate mitotic spindle disassembly and nuclear envelope sealing. Nature. 2015;522:231–5.
  43. Olmos Y, Hodgson L, Mantell J, Verkade P, Carlton JG. ESCRT-III controls nuclear envelope reformation. Nature. 2015;522:236–9.
  44. Lee C-P, Liu P-T, Kung H-N, Su M-T, Chua H-H, Chang Y-H, et al. The ESCRT machinery is recruited by the viral BFRF1 protein to the nucleus-associated membrane for the maturation of Epstein-Barr Virus. PLoS Pathog. 2012;8:e1002904.
  45. Webster BM, Colombi P, Jäger J, Lusk CP. Surveillance of nuclear pore complex assembly by ESCRT-III/Vps4. Cell. 2014;159:388–401.
  46. Raab M, Gentili M, de Belly H, Thiam HR, Vargas P, Jimenez AJ, et al. ESCRT III repairs nuclear envelope ruptures during cell migration to limit DNA damage and cell death. Science. 2016;352:359–362.
  47. Jimenez AJ, Maiuri P, Lafaurie-Janvore J, Divoux S, Piel M, Perez F. ESCRT machinery is required for plasma membrane repair. Science. 2014;343:1247136.
  48. Loncle N, Agromayor M, Martin-Serrano J, Williams DW. An ESCRT module is required for neuron pruning. Sci Rep. 2015;5:8461.
  49. Sweeney NT, Brenman JE, Jan YN, Gao F-B. The coiled-coil protein shrub controls neuronal morphogenesis in Drosophila. Curr Biol. 2006;16:1006–11.
  50. Zhang H, Wang Y, Wong JJL, Lim K-L, Liou Y-C, Wang H, Yu F. Endocytic pathways downregulate the L1-type cell adhesion molecule neuroglian to promote dendrite pruning in Drosophila. Dev Cell 2014;30:463–78.
  51. Issman-Zecharya N, Schuldiner O. The PI3K class III complex promotes axon pruning by downregulating a Ptc-derived signal via endosome-lysosomal degradation. Dev Cell. 2014;31:461–73.
  52. Frost A, Elgort MG, Brandman O, Ives C, Collins SR, Miller-Vedam L, et al. Functional repurposing revealed by comparing S. pombe and S. cerevisiae genetic interactions. Cell. 2012;149:1339–52.
  53. Morita E, Colf LA, Karren MA, Sandrin V, Rodesch CK, Sundquist WIHuman ESCRT-III and VPS4 proteins are required for centrosome and spindle maintenance. Proc Natl Acad Sci U S A. 2010;107:12889–94.
  54. Jin Y, Mancuso JJ, Uzawa S, Cronembold D, Cande WZ. The fission yeast homolog of the human transcription factor EAP30 blocks meiotic spindle pole body amplification. Dev Cell. 2005;9:63–73.
  55. Xie W, Li L, Cohen SN. Cell cycle-dependent subcellular localization of the TSG101 protein and mitotic and nuclear abnormalities associated with TSG101 deficiency. Proc Natl Acad Sci U S A. 1998;95:1595–600.
  56. Stauffer DR, Howard TL, Nyun T, Hollenberg SM. CHMP1 is a novel nuclear matrix protein affecting chromatin structure and cell-cycle progression. J Cell Sci. 2001;114:2383–93.
  57. Kamura T, Burian D, Khalili H, Schmidt SL, Sato S, Liu WJ, et al. Cloning and characterization of ELL-associated proteins EAP45 and EAP20. a role for yeast EAP-like proteins in regulation of gene expression by glucose. J Biol Chem. 2001;276:16528–33.
  58. Schmidt AE, Miller T, Schmidt SL, Shiekhattar R, Shilatifard A. Cloning and characterization of the EAP30 subunit of the ELL complex that confers derepression of transcription by RNA polymerase II. J Biol Chem. 1999;274:21981–5.
  59. Burgdorf S, Leister P, Scheidtmann KH. TSG101 interacts with apoptosis-antagonizing transcription factor and enhances androgen receptor-mediated transcription by promoting its monoubiquitination. J Biol Chem. 2004;279:17524–34.
  60. Sun Z, Pan J, Hope WX, Cohen SN, Balk SP. Tumor susceptibility gene 101 protein represses androgen receptor transactivation and interacts with p300. Cancer. 1999;86:689–96.
  61. Lin Y-S, Chen Y-J, Cohen SN, Cheng T-H. Identification of TSG101 functional domains and p21 loci required for TSG101-mediated p21 gene regulation. PLoS One. 2013;8:e79674.
  62. Irion U, St Johnston D. bicoid RNA localization requires specific binding of an endosomal sorting complex. Nature. 2007;445:554–8.
  63. Lee Y, Pressman S, Andress A, Kim K. Silencing by small RNAs is linked to endosomal trafficking. Nat Cell Biol. 2009;11:1150–6.
  64. Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat. Cell Biol. 2009;11:1143–9.
  65. Sigismund S, Confalonieri S, Ciliberto A, Polo S, Scita G, Di Fiore PP. Endocytosis and signaling: cell logistics shape the eukaryotic cell plan. Physiol Rev. 2012;92:273–366.
  66. Chanut-Delalande H, Jung AC, Baer MM, Lin L, Payre F, Affolter M. The Hrs/Stam complex acts as a positive and negative regulator of RTK signaling during Drosophila development. PLoS One. 2010;5:e10245.
  67. Lloyd TE, Atkinson R, Wu MN, Zhou Y, Pennetta G, Bellen HJ. Hrs Regulates Endosome Membrane Invagination and Tyrosine Kinase Receptor Signaling in Drosophila. Cell. 2002;108:261–9.
  68. Jékely G, Rørth P. Hrs mediates downregulation of multiple signalling receptors in Drosophila. EMBO Rep. 2003;4:1163–8.
  69. Bache KG, Stuffers S, Malerød L, Slagsvold T, Raiborg C, Lechardeur D, et al. The ESCRT-III subunit hVps24 is required for degradation but not silencing of the epidermal growth factor receptor. Mol Biol Cell. 2006;17:2513–23.
  70. 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. 2002;157:91–101.
  71. Babst M, Odorizzi G, Estepa EJ, Emr SD. Mammalian tumor susceptibility gene 101 (TSG101) and the yeast homologue, Vps23p, both function in late endosomal trafficking. Traffic. 2000;1:248–58.
  72. Bache KG, Slagsvold T, Cabezas A, Rosendal KR, Raiborg C, Stenmark H. The growth-regulatory protein HCRP1/hVps37A is a subunit of mammalian ESCRT-I and mediates receptor down-regulation. Mol Biol Cell. 2004;15: 4337–46.
  73. Doyotte A, Russell MRG, Hopkins CR, Woodman PG. Depletion of TSG101 forms a mammalian “Class E” compartment: a multicisternal early endosome with multiple sorting defects. J Cell Sci. 2005;118:3003–17.
  74. Baldys A, Raymond JR. Critical role of ESCRT machinery in EGFR recycling. Biochemistry. 2009;48:9321–3.
  75. Guruharsha KG, Kankel MW & Artavanis-Tsakonas S. The Notch signalling system: recent insights into the complexity of a conserved pathway. Nat Rev Genet. 2012;13:654–66.
  76. Moberg K, Schelble S, Burdick S, Hariharan I. Mutations in erupted, the Drosophila Ortholog of Mammalian Tumor Susceptibility Gene 101, Elicit Non-Cell-Autonomous Overgrowth. Dev Cell. 2005;9:699–710.
  77. Thompson BJ, Mathieu J, Sung H-H, Loeser E, Rørth P, Cohen SM. Tumor suppressor properties of the ESCRT-II complex component Vps25 in Drosophila. Dev Cell. 2005;9:711–20.
  78. Vaccari T, Bilder D. The Drosophila Tumor Suppressor vps25 Prevents Nonautonomous Overproliferation by Regulating Notch Trafficking. Dev Cell. 2005;9:687–98.
  79. Herz H-M, Chen Z, Scherr H, Lackey M, Bolduc C, Bergmann A. Vps25 Mosaics Display Non-Autonomous Cell Survival and Overgrowth, and Autonomous Apoptosis. Development. 2006;133:1871–80.
  80. Vaccari T, Rusten TE, Menut L, Nezis IP, Brech A, Stenmark H, Bilder D. Comparative analysis of ESCRT-I, ESCRT-II and ESCRT-III function in Drosophila by efficient isolation of ESCRT mutants. J Cell Sci. 2009;122:2413–23.
  81. Tognon E, Wollscheid N, Cortese K, Tacchetti C, Vaccari T. ESCRT-0 is not required for ectopic Notch activation and tumor suppression in Drosophila. PLoS One. 2014;9:e93987.
  82. Gilbert MM, Robinson BS, Moberg KH. Functional interactions between the erupted/tsg101 growth suppressor gene and the DaPKC and rbf1 genes in Drosophila imaginal disc tumors. PLoS One. 2009;4:e7039.
  83. Palacios F, Tushir JS, Fujita Y, D’Souza-Schorey C. Lysosomal targeting of E-cadherin: a unique mechanism for the down-regulation of cell-cell adhesion during epithelial to mesenchymal transitions. Mol Cell Biol. 2005;25:389–402.
  84. Dukes JD, Fish L, Richardson JD, Blaikley E, Burns S, Caunt CJ, et al. Functional ESCRT machinery is required for constitutive recycling of claudin-1 and maintenance of polarity in vertebrate epithelial cells. Mol Biol Cell. 2011;22:3192–205.
  85. Leithe E, Kjenseth A, Sirnes S, Stenmark H, Brech A, Rivedal E. Ubiquitylation of the gap junction protein connexin-43 signals its trafficking from early endosomes to lysosomes in a process mediated by Hrs and Tsg101. J Cell Sci. 2009;122:3883–93.
  86. Lobert VH, Stenmark H. Cell polarity and migration: emerging role for the endosomal sorting machinery. Physiology (Bethesda). 2011;26:171–80.
  87. Herz H-M, Woodfield SE, Chen Z, Bolduc C, Bergmann A. Common and distinct genetic properties of ESCRT-II components in Drosophila. PLoS One. 2009;4:e4165.
  88. Woodfield SE, Graves HK, Hernandez JA, Bergmann A. De-regulation of JNK and JAK/STAT signaling in ESCRT-II mutant tissues cooperatively contributes to neoplastic tumorigenesis. PLoS One. 2013;8:e56021.
  89. Vaccari T, Bilder D. At the crossroads of polarity, proliferation and apoptosis: the use of Drosophila to unravel the multifaceted role of endocytosis in tumor suppression. Mol Oncol. 2009;3:354–65.
  90. Komada M, Soriano P. Hrs, a FYVE finger protein localized to early endosomes, is implicated in vesicular traffic and required for ventral folding morphogenesis. Genes Dev. 1999;13:1475–85.
  91. Ruland J, Sirard C, Elia A, MacPherson D, Wakeham A, Li L, et al. p53 accumulation, defective cell proliferation, and early embryonic lethality in mice lacking tsg101. Proc Natl Acad Sci U S A. 2001;98:1859–64.
  92. Yamada M, Ishii N, Asao H, Murata K, Kanazawa C, Sasaki H, Sugamura K. Signal-transducing adaptor molecules STAM1 and STAM2 are required for T-cell development and survival. Mol Cell Biol. 2002;22:8648–58.
  93. Shim J-H, Xiao C, Hayden MS, Lee K-Y, Trombetta ES, Pypaert M, et al. CHMP5 is essential for late endosome function and down-regulation of receptor signaling during mouse embryogenesis. J Cell Biol. 2006;172:1045–56.
  94. Handschuh K, Feenstra J, Koss M, Ferretti E, Risolino M, Zewdu R, et al. ESCRT-II/Vps25 constrains digit number by endosome-mediated selective modulation of FGF-SHH signaling. Cell Rep. 2014;9:674–87.
  95. Rodahl LM, Haglund K, Sem-Jacobsen C, Wendler F, Vincent J-P, Lindmo K, et al. Disruption of Vps4 and JNK function in Drosophila causes tumour growth. PLoS One. 2009;4:e4354.
  96. Huang H-R, Chen ZJ, Kunes S, Chang G-D, Maniatis T. Endocytic pathway is required for Drosophila Toll innate immune signaling. Proc Natl Acad Sci U S A. 2010;107:8322–7.
  97. Seto ES, Bellen HJ. Internalization is required for proper Wingless signaling in Drosophila melanogaster. J. Cell Biol. 2006;173:95–106.
  98. Lund VK, Delotto R. Regulation of Toll and Toll-like receptor signaling by the endocytic pathway. Small GTPases 2011;2:95–8.
  99. Matusek T, Wendler F, Polès S, Pizette S, D’Angelo G, Fürthauer M, Thérond PP. The ESCRT machinery regulates the secretion and long-range activity of Hedgehog. Nature. 2014;516:99–103.
  100. Taelman VF, Dobrowolski R, Plouhinec J-L, Fuentealba LC, Vorwald PP, Gumper I, et al. Wnt signaling requires sequestration of glycogen synthase kinase 3 inside multivesicular endosomes. Cell. 2010;143:1136–48.
  101. Mamińska A, Bartosik A, Banach-Orłowska M, Pilecka I, Jastrzębski K, Zdżalik-Bielecka D, et al. ESCRT proteins restrict constitutive NF-κB signaling by trafficking cytokine receptors. Sci Signal. 2016;9:ra8-ra8.
  102. Corless L, Crump CM, Griffin SDC, Harris M. Vps4 and the ESCRT-III complex are required for the release of infectious hepatitis C virus particles. J Gen Virol. 2010;91:362–72.
  103. Jun M-H, Han J-H, Lee Y-K, Jang D-J, Kaang B-K, Lee J-A. TMEM106B, a frontotemporal lobar dementia (FTLD) modifier, associates with FTD-3-linked CHMP2B, a complex of ESCRT-III. Mol Brain. 2015;8:85.
  104. Effantin G, Dordor A, Sandrin V, Martinelli N, Sundquist WI, Schoehn G, Weissenhorn W. ESCRT-III CHMP2A and CHMP3 form variable helical polymers in vitro and act synergistically during HIV-1 budding. Cell Microbiol. 2013;15:213–26.
  105. Bleck M, Itano MS, Johnson DS, Thomas VK, North AJ, Bieniasz PD, Simon SM. Temporal and spatial organization of ESCRT protein recruitment during HIV-1 budding. Proc Natl Acad Sci U S A. 2014;111:12211–6.
  106. Van Engelenburg SB, Shtengel G, Sengupta P, Waki K, Jarnik M, Ablan SD, et al. Distribution of ESCRT machinery at HIV assembly sites reveals virus scaffolding of ESCRT subunits. Science. 2014;343:653–6.
  107. Sandrin V, Sundquist WI. ESCRT requirements for EIAV budding. Retrovirology. 2013;10:104.
  108. Prescher J, Baumgärtel V, Ivanchenko S, Torrano AA, Bräuchle C, Müller B, Lamb DCSuper-resolution imaging of ESCRT-proteins at HIV-1 assembly sites. PLoS Pathog. 2015;11:e1004677.
  109. Morita E, Sandrin V, McCullough J, Katsuyama A, Baci Hamilton I, Sundquist WI. ESCRT-III protein requirements for HIV-1 budding. Cell Host Microbe. 2011;9:235–42.
  110. Pawliczek T, Crump CM. Herpes simplex virus type 1 production requires a functional ESCRT-III complex but is independent of TSG101 and ALIX expression. J Virol. 2009;83:11254–64.
  111. Silvestri LS, Ruthel G, Kallstrom G, Warfield KL, Swenson DL, Nelle T, et al. Involvement of vacuolar protein sorting pathway in Ebola virus release independent of TSG101 interaction. J Infect Dis. 2007;196(Suppl):S264–70.
  112. Madison MN, Okeoma CM. Exosomes: Implications in HIV-1 Pathogenesis. Viruses. 2015;7:4093–118.
  113. Narayanan A, Iordanskiy S, Das R, Van Duyne R, Santos S, Jaworski E, et al. Exosomes derived from HIV-1-infected cells contain trans-activation response element RNA. J Biol Chem. 2013;288:20014–33.
  114. Kadiu I, Narayanasamy P, Dash PK, Zhang W, Gendelman HE. Biochemical and biologic characterization of exosomes and microvesicles as facilitators of HIV-1 infection in macrophages. J Immunol. 2012;189:744–54.
  115. Tamai K, Shiina M, Tanaka N, Nakano T, Yamamoto A, Kondo Y, et al. Regulation of hepatitis C virus secretion by the Hrs-dependent exosomal pathway. Virology. 2012;422:377–85.
  116. Temme S, Eis-Hübinger AM, McLellan AD, Koch N. The herpes simplex virus-1 encoded glycoprotein B diverts HLA-DR into the exosome pathway. J Immunol. 2010;184:236–43.
  117. Alenquer M, Amorim M. Exosome Biogenesis, Regulation, and Function in Viral Infection. Viruses. 2015;7:5066–83.
  118. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MAJ, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci U S A. 2010;107:6328–33.
  119. Meckes DG, Shair KHY, Marquitz AR, Kung C-P, Edwards RH, Raab-Traub N. Human tumor virus utilizes exosomes for intercellular communication. Proc Natl Acad Sci U S A. 2010;10:20370–5.
  120. Silva-Ayala D, López T, Gutiérrez M, Perrimon N, López S, Arias CF. Genome-wide RNAi screen reveals a role for the ESCRT complex in rotavirus cell entry. Proc Natl Acad Sci U S A. 2013;110:10270–5.
  121. Li Z, Blissard GW. Cellular VPS4 is required for efficient entry and egress of budded virions of Autographa californica multiple nucleopolyhedrovirus. J Virol. 2012;86:459–72.
  122. Shtanko O, Nikitina RA, Altuntas CZ, Chepurnov AA, Davey RA. Crimean-Congo hemorrhagic fever virus entry into host cells occurs through the multivesicular body and requires ESCRT regulators. PLoS Pathog. 2014;10:e1004390.
  123. Pasqual G, Rojek JM, Masin M, Chatton J-Y, Kunz S. Old world arenaviruses enter the host cell via the multivesicular body and depend on the endosomal sorting complex required for transport. PLoS Pathog. 2011;7:e1002232.
  124. Broniarczyk J, Bergant M, Goździcka-Józefiak A, Banks L. Human papillomavirus infection requires the TSG101 component of the ESCRT machinery. Virology. 2014;460–461, 83–90.
  125. Garrison AR, Radoshitzky SR, Kota KP, Pegoraro G, Ruthel G, Kuhn JH, et al. Crimean-Congo hemorrhagic fever virus utilizes a clathrin- and early endosome-dependent entry pathway. Virology. 2013;444:45–54.
  126. Simon M, Johansson C, Mirazimi A. Crimean-Congo hemorrhagic fever virus entry and replication is clathrin-, pH- and cholesterol-dependent. J Gen Virol. 2009;90:210–5.
  127. Ghoujal B, Milev MP, Ajamian L, Abel K, Mouland AJ. ESCRT-II’s involvement in HIV-1 genomic RNA trafficking and assembly. Biol Cell. 2012;104:706–21.
  128. Meng B, Ip NCY, Prestwood LJ, Abbink TEM, Lever AML. Evidence that the endosomal sorting complex required for transport-II (ESCRT-II) is required for efficient human immunodeficiency virus-1 (HIV-1) production. Retrovirology. 2015;12:72.
  129. Stieler JT, Prange R. Involvement of ESCRT-II in hepatitis B virus morphogenesis. PLoS One. 2014;9:e91279.
  130. Agaisse H, Burrack LS, Philips JA, Rubin EJ, Perrimon N, Higgins DE. Genome-wide RNAi screen for host factors required for intracellular bacterial infection. Science. 2005;309:1248–51.
  131. Philips JA, Porto MC, Wang H, Rubin EJ, Perrimon N. ESCRT factors restrict mycobacterial growth. Proc Natl Acad Sci U S A. 2008;105:3070–5.
  132. Mehra A, Zahra A, Thompson V, Sirisaengtaksin N, Wells A, Porto M, et al. Mycobacterium tuberculosis Type VII Secreted Effector EsxH Targets Host ESCRT to Impair Trafficking. PLoS Pathog. 2013;9:e1003734.
  133. Abrami L, Brandi L, Moayeri M, Brown MJ, Krantz BA, Leppla SH, VanderGoot FG. Hijacking Multivesicular Bodies Enables Long-Term and Exosome-Mediated Long-Distance Action of Anthrax Toxin. Cell Rep. 2013;5:986–96.
  134. Wolf JM, Johnson DJ, Chmielewski D, Davis DA. The Candida albicans ESCRT pathway makes Rim101-dependent and -independent contributions to pathogenesis. Eukaryot. Cell. 2010;9:1203–15.
  135. Cornet M, Bidard F, Schwarz P, Da Costa G, Blanchin-Roland S, Dromer F, Gaillardin C. Deletions of endocytic components VPS28 and VPS32 affect growth at alkaline pH and virulence through both RIM101-dependent and RIM101-independent pathways in Candida albicans. Infect Immun. 2005;73:7977–87.
  136. Kullas AL, Li M, Davis DA. Snf7p, a component of the ESCRT-III protein complex, is an upstream member of the RIM101 pathway in Candida albicans. Eukaryot Cell. 2004;3:1609–18.
  137. Xu W, Smith FJ, Subaran R, Mitchell AP. Multivesicular body-ESCRT components function in pH response regulation in Saccharomyces cerevisiae and Candida albicans. Mol Biol Cell. 2004;15:5528–37.
  138. Zhang Y, Li W, Chu M, Chen H, Yu H, Fang C, et al. The AAA ATPase Vps4 Plays Important Roles in Candida albicans Hyphal Formation and is Inhibited by DBeQ. Mycopathologia. 2015;181:1–11.
  139. Wolf JM, Davis DA. Mutational analysis of Candida albicans SNF7 reveals genetically separable Rim101 and ESCRT functions and demonstrates divergence in bro1-domain protein interactions. Genetics. 2010;184:673–94.
  140. Li L, Cohen SN. tsg101: A Novel Tumor Susceptibility Gene Isolated by Controlled Homozygous Functional Knockout of Allelic Loci in Mammalian Cells. Cell. 1996;85:319–29.
  141. Broniarczyk J, Olejnik-Schmidt AK, Luczak MW, Schmidt MT, Dabrowski M, Józefiak A, et al. Analysis of expression and structure of the TSG101 gene in cervical cancer cells. Int J Mol Med. 2010;25:777–83.
  142. Krempler A, Henry MD, Triplett AA, Wagner K-U. Targeted deletion of the Tsg101 gene results in cell cycle arrest at G1/S and p53-independent cell death. J Biol Chem. 2002;277:43216–23.
  143. Wagner K-U, Krempler A, Qi Y, Park K, Henry MD, Triplett AA, et al. Tsg101 is essential for cell growth, proliferation, and cell survival of embryonic and adult tissues. Mol Cell Biol. 2003;23:150–62.
  144. Feng GH, Lih CJ, Cohen SN. TSG101 protein steady-state level is regulated posttranslationally by an evolutionarily conserved COOH-terminal sequence. Cancer Res. 2000;60:1736–41.
  145. Oh KB, Stanton MJ, West WW, Todd GL, Wagner K-U. Tsg101 is upregulated in a subset of invasive human breast cancers and its targeted overexpression in transgenic mice reveals weak oncogenic properties for mammary cancer initiation. Oncogene. 2007;26:5950–9.
  146. Liu F, Yu Y, Jin Y, Fu S. TSG101, identified by screening a cancer cDNA library and soft agar assay, promotes cell proliferation in human lung cancer. Mol Biol Rep. 2010;37:2829–38.
  147. Liu D, Yang Z, Jiang S. Identification of PEG10 and TSG101 as carcinogenesis, progression, and poor-prognosis related biomarkers for gallbladder adenocarcinoma. Pathol Oncol Res. 2011;17:859–66.
  148. Liu R-T, Huang C-C, You H-L, Chou F-F, Hu C-CA, Chao F-P, et al. Overexpression of tumor susceptibility gene TSG101 in human papillary thyroid carcinomas. Oncogene. 2002;21:4830–7.
  149. Young TW, Mei FC, Rosen DG, Yang G, Li N, Liu J, Cheng X. Up-regulation of tumor susceptibility gene 101 protein in ovarian carcinomas revealed by proteomics analyses. Mol Cell Proteomics. 2007;6:294–304.
  150. Zhui G, Gilchrist R, Borley N, Chng HW, Morgan M, Marshall JF, et al. Reduction of TSG101 protein has a negative impact on tumor cell growth. Int J Cancer. 2004;109:541–7.
  151. Zhang Y, Song M, Cui ZS, Li CY, Xue XX, Yu M, Lu Y, et al. Down-regulation of TSG101 by small interfering RNA inhibits the proliferation of breast cancer cells through the MAPK/ERK signal pathway. Histol Histopathol. 2011;26:87–94.
  152. Young TW, Rosen DG, Mei FC, Li N, Liu J, Wang X-F, Cheng X. Up-regulation of tumor susceptibility gene 101 conveys poor prognosis through suppression of p21 expression in ovarian cancer. Clin Cancer Res. 2007;13:3848–54.
  153. Xu Z, Liang L, Wang H, Li T, Zhao M. HCRP1, a novel gene that is downregulated in hepatocellular carcinoma, encodes a growth-inhibitory protein. Biochem Biophys Res Commun. 2003;311:1057–66.
  154. Lai MW, Huang SF, Lin SM, Chen TC, Lin CY, Yeh CN, et al. Expression of the HCRP1 mRNA in HCC as an independent predictor of disease-free survival after surgical resection. Hepatol Res. 2009;39:164–76.
  155. Xu J, Yang W, Wang Q, Zhang Q, Li X, Lin X, et al. Decreased HCRP1 expression is associated with poor prognosis in breast cancer patients. Int J Clin Exp Pathol. 2014;7:7915–22.
  156. Wittinger M, Vanhara P, El-Gazzar A, Savarese-Brenner B, Pils D, Anees M, et al. hVps37A Status affects prognosis and cetuximab sensitivity in ovarian cancer. Clin Cancer Res. 2011;17:7816–27.
  157. Chen F, Deng J, Liu X, Li W, Zheng J. HCRP-1 regulates cell migration and invasion via EGFR-ERK mediated up-regulation of MMP-2 with prognostic significance in human renal cell carcinoma. Sci Rep. 2015;5:13470.
  158. You Z, Xin Y, Liu Y, Sun J, Zhou G, Gao H, Xu P, et al. Chmp1A acts as a tumor suppressor gene that inhibits proliferation of renal cell carcinoma. Cancer Lett. 2012;319:190–6.
  159. Li J, Belogortseva N, Porter D, Park M. Chmp1A functions as a novel tumor suppressor gene in human embryonic kidney and ductal pancreatic tumor cells. Cell Cycle. 2008;7:2886–93.
  160. Mochida GH, Ganesh VS, de Michelena MI, Dias H, Atabay KD, Kathrein KL, et al. CHMP1A encodes an essential regulator of BMI1-INK4A in cerebellar development. Nat Genet. 2012;44:1260–4.
  161. Hu B, Jiang D, Chen Y, Wei L, Zhang S, Zhao F, Ni R, et al. High CHMP4B expression is associated with accelerated cell proliferation and resistance to doxorubicin in hepatocellular carcinoma. Tumor Biol. 2015;36:2569–81.
  162. Li K, Liu J, Tian M, Gao G, Qi X, Pan Y, et al. CHMP4C Disruption Sensitizes the Human Lung Cancer Cells to Irradiation. Int J Mol Sci. 2015;17.
  163. Wei J, Lv L, Wan Y, Cao Y, Li G, Lin H, et al. Vps4A functions as a tumor suppressor by regulating the secretion and uptake of exosomal microRNAs in human hepatoma cells. Hepatology. 2015;61:1284–94.
  164. Chen VY, Posada MM, Blazer LL, Zhao T, Rosania GR. The role of the VPS4A-exosome pathway in the intrinsic egress route of a DNA-binding anticancer drug. Pharm Res. 2006;23:1687–95.
  165. Safaei R, Larson BJ, Cheng TC, Gibson MA, Otani S, Naerdemann W, Howell SB. Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol Cancer Ther. 2005;4:1595–604.
  166. Toyoshima M, Tanaka N, Aoki J, Tanaka Y, Murata K, Kyuuma M, et al. Inhibition of tumor growth and metastasis by depletion of vesicular sorting protein Hrs: Its regulatory role on E-cadherin and β-catenin. Cancer Res. 2007;67:5162–71.
  167. Skibinski G, Parkinson NJ, Brown JM, Chakrabarti L, Lloyd SL, Hummerich H, et al. Mutations in the endosomal ESCRTIII-complex subunit CHMP2B in frontotemporal dementia. Nat Genet. 2005;37:806–8.
  168. van der Zee J, Urwin H, Engelborghs S, Bruyland M, Vandenberghe R, Dermaut B, et al. CHMP2B C-truncating mutations in frontotemporal lobar degeneration are associated with an aberrant endosomal phenotype in vitro. Hum Mol Genet. 2008;17:313–22.
  169. Zamborlini A, Usami Y, Radoshitzky SR, Popova E, Palu G, Göttlinger H. Release of autoinhibition converts ESCRT-III components into potent inhibitors of HIV-1 budding. Proc Natl Acad Sci U S A. 2006;103:19140–5.
  170. Stuchell-Brereton MD, Skalicky JJ, Kieffer C, Karren MA, Ghaffarian S, Sundquist WI. ESCRT-III recognition by VPS4 ATPases. Nature. 2007;449:740–4.
  171. Obita T, Saksena S, Ghazi-Tabatabai S, Gill DJ, Perisic O, Emr SD, Williams RL. Structural basis for selective recognition of ESCRT-III by the AAA ATPase Vps4. Nature. 2007;449:735–9.
  172. Urwin H, Authier A, Nielsen JE, Metcalf D, Powell C, Froud K, et al. Disruption of endocytic trafficking in frontotemporal dementia with CHMP2B mutations. Hum Mol Genet. 2010;19:2228–38.
  173. Lee J-A, Beigneux A, Ahmad ST, Young SG, Gao F-B. ESCRT-III dysfunction causes autophagosome accumulation and neurodegeneration. Curr Biol. 2007;17:1561–7.
  174. Nielsen TT, Mizielinska S, Hasholt L, Isaacs AM, Nielsen JE. Reversal of pathology in CHMP2B-mediated frontotemporal dementia patient cells using RNA interference. J Gene Med. 2012;14:521–9.
  175. Lee YS, Dutta A. MicroRNAs in cancer. Annu Rev Pathol. 2009;4:199–227.
  176. Cox LE, Ferraiuolo L, Goodall EF, Heath PR, Higginbottom A, Mortiboys H, et al. Mutations in CHMP2B in lower motor neuron predominant amyotrophic lateral sclerosis (ALS). PLoS One. 2010;5:e9872.
  177. Holm IE, Englund E, Mackenzie IRA, Johannsen P, Isaacs AM. A reassessment of the neuropathology of frontotemporal dementia linked to chromosome 3. J Neuropathol Exp Neurol. 2007;66:884–91.
  178. Parkinson N, Ince PG, Smith MO, Highley R, Skibinski G, Andersen PM, et al. ALS phenotypes with mutations in CHMP2B (charged multivesicular body protein 2B). Neurology. 2006;67:1074–7.
  179. Reid E, Connell J, Edwards TL, Duley S, Brown SE, Sanderson CM. The hereditary spastic paraplegia protein spastin interacts with the ESCRT-III complex-associated endosomal protein CHMP1B. Hum Mol Genet. 2005;14:19–38.
  180. Park SH, Zhu P-P, Parker RL, Blackstone C. Hereditary spastic paraplegia proteins REEP1, spastin, and atlastin-1 coordinate microtubule interactions with the tubular ER network. J Clin Invest. 2010;120:1097–110.
  181. Papadopoulos C, Orso G, Mancuso G, Herholz M, Gumeni S, Tadepalle N, et al. Spastin binds to lipid droplets and affects lipid metabolism. PLoS Genet. 2015;11:e1005149.
  182. Zivony-Elboum Y, Westbroek W, Kfir N, Savitzki D, Shoval Y, Bloom A, et al. A founder mutation in Vps37A causes autosomal recessive complex hereditary spastic paraparesis. J Med Genet. 2012;49:462–72.
  183. Baba M, Nakajo S, Tu PH, Tomita T, Nakaya K, Lee VM, Trojanowski JQ, Iwatsubo T. Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies. Am J Pathol. 1998;152:879–84.
  184. Braak H, Tredici K Del, Rüb U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.
  185. Lee S-J, Lim H-S, Masliah E, Lee H-J. Protein aggregate spreading in neurodegenerative diseases: problems and perspectives. Neurosci Res. 2011;70:339–48.
  186. Spencer B, Kim C, Gonzalez T, Bisquertt A, Patrick C, Rockenstein E, et al. α-Synuclein interferes with the ESCRT-III complex contributing to the pathogenesis of Lewy Body disease. Hum Mol Genet. 2016;25:1100–15.
  187. Hasegawa T, Konno M, Baba T, Sugeno N, Kikuchi A, Kobayashi M, et al. The AAA-ATPase VPS4 regulates extracellular secretion and lysosomal targeting of α-synuclein. PLoS One. 2011;6:e29460.
  188. Alvarez-Erviti L, Seow Y, Schapira AH, Gardiner C, Sargent IL, Wood MJA, Cooper JM. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol Dis. 2011;42:360–7.
  189. Spencer B, Emadi S, Desplats P, Eleuteri S, Michael S, Kosberg K, et al. ESCRT-mediated uptake and degradation of brain-targeted α-synuclein single chain antibody attenuates neuronal degeneration in vivo. Mol Ther. 2014;22:1753–67.
  190. Kurashige T, Takahashi T, Yamazaki Y, Hiji M, Izumi Y, Yamawaki T, Matsumoto M. Localization of CHMP2B-immunoreactivity in the brainstem of Lewy body disease. Neuropathology. 2013;33:237–45.
  191. Edgar JR, Willén K, Gouras GK, Futter CE. ESCRTs regulate amyloid precursor protein sorting in multivesicular bodies and intracellular amyloid-β accumulation. J Cell Sci. 2015;128:2520–8.
  192. Filimonenko M, Stuffers S, Raiborg C, Yamamoto A, Malerød L, Fisher EMC, et al. Functional multivesicular bodies are required for autophagic clearance of protein aggregates associated with neurodegenerative disease. J Cell Biol. 2007;179:485–500.
  193. Yamamoto A, Cremona ML, Rothman JE. Autophagy-mediated clearance of huntingtin aggregates triggered by the insulin-signaling pathway. J Cell Biol. 2006;172:719–31.
  194. Shiels A, Bennett TM, Knopf HLS, Yamada K, Yoshiura K, Niikawa N, Shim S, Hanson PI. CHMP4B, a novel gene for autosomal dominant cataracts linked to chromosome 20q. Am J Hum Genet. 2007;81:596–606.
  195. Sagona AP, Nezis IP, Stenmark H. Association of CHMP4B and autophagy with micronuclei: implications for cataract formation. Biomed Res Int. 2014, 974393.
  196. Zhang D, Wang L, Yan L, Miao X, Gong C, Xiao M, et al. Vacuolar protein sorting 4B regulates apoptosis of intestinal epithelial cells via p38 MAPK in Crohn’s disease. Exp Mol Pathol. 2015;98:55–64.
  197. Hurley JH. ESCRTs are everywhere. EMBO J. 2015;34:2398–407.
  198. 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. 1995;15:6213–21.
  199. Lu L, Komada M, Kitamura N. Human Hrs, a tyrosine kinase substrate in growth factor-stimulated cells: cDNA cloning and mapping of the gene to chromosome 17. Gene. 1998;213:125–32.
  200. Urbé S, Mills IG, Stenmark H, Kitamura N, Clague MJ. Endosomal localization and receptor dynamics determine tyrosine phosphorylation of hepatocyte growth factor-regulated tyrosine kinase substrate. Mol Cell Biol. 2000;20:7685–92.
  201. Stern KA, Visser Smit GD, Place TL, Winistorfer S, Piper RC, Lill NL. Epidermal growth factor receptor fate is controlled by Hrs tyrosine phosphorylation sites that regulate Hrs degradation. Mol Cell Biol. 2007;27:888–98.
  202. Urbé S, Sachse M, Row PE, Preisinger C, Barr FA, Strous G, Klumperman J, Clague MJ. The UIM domain of Hrs couples receptor sorting to vesicle formation. J Cell Sci. 2003;116:4169–79.
  203. Hoeller D, Crosetto N, Blagoev B, Raiborg C, Tikkanen R, Wagner S, et al. Regulation of ubiquitin-binding proteins by monoubiquitination. Nat Cell Biol. 2006;8:163–9.
  204. Row PE, Clague MJ, Urbé S. Growth factors induce differential phosphorylation profiles of the Hrs-STAM complex: a common node in signalling networks with signal-specific properties. Biochem J. 2005;389:629–36.
  205. Kim BY, Olzmann JA, Barsh GS, Chin L-S, Li L. Spongiform neurodegeneration-associated E3 ligase Mahogunin ubiquitylates TSG101 and regulates endosomal trafficking. Mol Biol Cell. 2007;18:1129–42.
  206. Jiao J, Sun K, Walker WP, Bagher P, Cota CD, Gunn TM. Abnormal regulation of TSG101 in mice with spongiform neurodegeneration. Biochim Biophys Acta. 2009;1792:1027–35.
  207. Majumder P, Chakrabarti O. Mahogunin regulates fusion between amphisomes/MVBs and lysosomes via ubiquitination of TSG101. Cell Death Dis. 2015;6:e1970.
  208. Amit I, Yakir L, Katz M, Zwang Y, Marmor MD, Citri A, et al. Tal, a Tsg101-specific E3 ubiquitin ligase, regulates receptor endocytosis and retrovirus budding. Genes Dev. 2004;18:1737–52.
  209. McDonald B, Martin-Serrano J. Regulation of Tsg101 expression by the steadiness box: a role of Tsg101-associated ligase. Mol Biol Cell. 2008;19:754–63.
  210. Martinelli N, Hartlieb B, Usami Y, Sabin C, Dordor A, Miguet N, et al. CC2D1A is a regulator of ESCRT-III CHMP4B. J Mol Biol. 2012;419:75–88.
  211. Usami Y, Popov S, Weiss ER, Vriesema-Magnuson C, Calistri A, Göttlinger HG. Regulation of CHMP4/ESCRT-III function in human immunodeficiency virus type 1 budding by CC2D1A. J Virol. 2012;86:3746–56.
  212. >Drusenheimer N, Migdal B, Jäckel S, Tveriakhina L, Scheider K, Schulz K, et al. The Mammalian Orthologs of Drosophila Lgd, CC2D1A and CC2D1B, Function in the Endocytic Pathway, but Their Individual Loss of Function Does Not Affect Notch Signalling. PLoS Genet. 2015;11:e1005749.
  213. Jaekel R, Klein T. The Drosophila Notch inhibitor and tumor suppressor gene lethal (2) giant discs encodes a conserved regulator of endosomal trafficking. Dev Cell. 2006;11:655–69.
  214. Troost T, Jaeckel S, Ohlenhard N, Klein T. The tumour suppressor Lethal (2) giant discs is required for the function of the ESCRT-III component Shrub/CHMP4. J Cell Sci. 2012;125:763–76.
  215. Gallagher CM, Knoblich JA. The conserved c2 domain protein lethal (2) giant discs regulates protein trafficking in Drosophila. Dev Cell. 2006;11:641–53.
  216. Matias NR, Mathieu J, Huynh J-R. Abscission is regulated by the ESCRT-III protein shrub in Drosophila germline stem cells. PLoS Genet. 2015;11:e1004653.
  217. Morawa KS, Schneider M, Klein T. Lgd regulates the activity of the BMP/Dpp signalling pathway during Drosophila oogenesis. Development. 2015;142:1325–35.
  218. Roy M, Pear WS, Aster JC. The multifaceted role of Notch in cancer. Curr Opin Genet Dev. 2007;17:52–9.
  219. Palmer WH, Deng W-M. Ligand-Independent Mechanisms of Notch Activity. Trends Cell Biol. 2015;25:697–707.
  220. Kobia F, Duchi S, Deflorian G, Vaccari T. Pharmacologic inhibition of vacuolar H+ ATPase reduces physiologic and oncogenic Notch signaling. Mol Oncol. 2014;8:207–20.
  221. Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, Erdin S, et al. TFEB links autophagy to lysosomal biogenesis. Science. 2011;332:1429–33.
  222. Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y, et al. An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Chem. 2009;284:8023–32.
  223. Settembre C, Zoncu R, Medina DL, Vetrini F, Erdin S, Erdin S, et al. A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. EMBO J. 2012;31:1095–108.
  224. Roczniak-Ferguson A, Petit CS, Froehlich F, Qian S, Ky J, Angarola B, et al. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci Signal. 2012;5:ra42.
  225. Song W, Wang F, Lotfi P, Sardiello M, Segatori L. 2-Hydroxypropyl-β-cyclodextrin promotes transcription factor EB-mediated activation of autophagy: Implications for therapy. J Biol Chem. 2014;289:10211–22.
  226. Decressac M, Mattsson B, Weikop P, Lundblad M, Jakobsson J, Björklund A. TFEB-mediated autophagy rescues midbrain dopamine neurons from α-synuclein toxicity. Proc Natl Acad Sci U S A. 2013;110:E1817-26.
  227. Bové J, Martínez-Vicente M, Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci. 2011;12,:437–52.
  228. Kilpatrick K, Zeng Y, Hancock T, Segatori L. Genetic and chemical activation of TFEB mediates clearance of aggregated α-synuclein. PLoS One. 2015;10:e0120819.
  229. Tognon E, Kobia F, Busi I, Fumagalli A, De Masi F, Vaccari T. Control of lysosomal biogenesis and Notch-dependent tissue patterning by components of the TFEB-V-ATPase axis in Drosophila melanogaster. Autophagy. 2016;12:499–514.
  230. Miura GI, Roignant J-Y, Wassef M, Treisman JE. Myopic acts in the endocytic pathway to enhance signaling by the Drosophila EGF receptor. Development. 2008;135:1913–22.
  231. Parkinson MDJ, Piper SC, Bright NA, Evans JL, Boname JM, Bowers K, et al. A non-canonical ESCRT pathway, including histidine domain phosphotyrosine phosphatase (HD-PTP), is used for down-regulation of virally ubiquitinated MHC class I. Biochem J. 2015;471:79–88.
  232. Ali N, Zhang L, Taylor S, Mironov A, Urbé S, Woodman P. Recruitment of UBPY and ESCRT exchange drive HD-PTP-dependent sorting of EGFR to the MVB. Curr Biol. 2013;23:453–61.
  233. Lee J, Oh K-J, Lee D, Kim BY, Choi JS, Ku B, Kim SJ. Structural Study of the HD-PTP Bro1 Domain in a Complex with the Core Region of STAM2, a Subunit of ESCRT-0. PLoS One. 2016;11:e0149113.
  234. Pradhan-Sundd T, Verheyen EM. The Myopic-Ubpy-Hrs nexus enables endosomal recycling of Frizzled. Mol Biol Cell. 2015;26:3329–42.
  235. Toyooka S, Ouchida M, Jitsumori Y, Tsukuda K, Sakai A, Nakamura A, et al. HD-PTP: A novel protein tyrosine phosphatase gene on human chromosome 3p21.3. Biochem Biophys Res Commun. 2000;278:671–8.
  236. Gingras M-C, Zhang YL, Kharitidi D, Barr AJ, Knapp S, Tremblay ML, Pause A. HD-PTP is a catalytically inactive tyrosine phosphatase due to a conserved divergence in its phosphatase domain. PLoS One. 2009;4:e5105.
  237. Haller O, Weber F. Pathogenic viruses: smart manipulators of the interferon system. Curr Top Microbio. Immunol. 2007;316:315–34.
  238. Harty RN, Pitha PM, Okumura A. Antiviral activity of innate immune protein ISG15. J Innate Immun. 2009;1:397–404.
  239. Morales DJ, Lenschow DJ. The antiviral activities of ISG15. J Mol Biol. 2013;425:4995–5008.
  240. Lenschow DJ, Lai C, Frias-Staheli N, Giannakopoulos N V, Lutz A, Wolff T, et al. IFN-stimulated gene 15 functions as a critical antiviral molecule against influenza, herpes, and Sindbis viruses. Proc Natl Acad Sci U S A. 2007;104:1371–6.
  241. Okumura A, Lu G, Pitha-Rowe I, Pitha PM. Innate antiviral response targets HIV-1 release by the induction of ubiquitin-like protein ISG15. Proc Natl Acad Sci U S A. 2006;103:1440–5.
  242. Pincetic A, Kuang Z, Seo EJ, Leis J. The interferon-induced gene ISG15 blocks retrovirus release from cells late in the budding process. J Virol. 2010;84:4725–36.
  243. Vild CJ, Li Y, Guo EZ, Liu Y, Xu Z. A novel mechanism of regulating the ATPase VPS4 by its cofactor LIP5 and the endosomal sorting complex required for transport (ESCRT)-III protein CHMP5. J Biol Chem. 2015;290:7291–303.
  244. Shim S, Merrill SA, Hanson PI. Novel interactions of ESCRT-III with LIP5 and VPS4 and their implications for ESCRT-III disassembly. Mol Biol Cell. 2008;19:2661–72.
  245. Skalicky JJ, Arii J, Wenzel DM, Stubblefield W-MB, Katsuyama A, Uter NT, et al. Interactions of the human LIP5 regulatory protein with endosomal sorting complexes required for transport. J Biol Chem. 2012;287:43910–26.
  246. Goila-Gaur R, Demirov DG, Orenstein JM, Ono A, Freed EO. Defects in human immunodeficiency virus budding and endosomal sorting induced by TSG101 overexpression. J Virol. 2003;77:6507–19.
  247. Demirov DG, Ono A, Orenstein JM, Freed EO. Overexpression of the N-terminal domain of TSG101 inhibits HIV-1 budding by blocking late domain function. Proc Natl Acad Sci U S A. 2002;99:955–60.
  248. Chen H, Liu X, Li Z, Zhan P, De Clercq E. TSG101: a novel anti-HIV-1 drug target. Curr Med Chem. 2010;17:750–8.
  249. Madara JJ, Han Z, Ruthel G, Freedman BD, Harty RN. The multifunctional Ebola virus VP40 matrix protein is a promising therapeutic target. Future Virol. 2015;10:537–546.
  250. Tavassoli A, Lu Q, Gam J, Pan H, Benkovic SJ, Cohen SN. Inhibition of HIV budding by a genetically selected cyclic peptide targeting the Gag-TSG101 interaction. ACS Chem Biol. 2008;3:757–64.
  251. Martin-Serrano J, Neil SJD. Host factors involved in retroviral budding and release. Nat Rev Microbiol. 2011;9:519–31.