101
|
Connell JW, Allison RJ, Rodger CE, Pearson G, Zlamalova E, Reid E. ESCRT-III-associated proteins and spastin inhibit protrudin-dependent polarised membrane traffic. Cell Mol Life Sci 2020; 77:2641-2658. [PMID: 31587092 PMCID: PMC7320071 DOI: 10.1007/s00018-019-03313-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/27/2019] [Accepted: 09/19/2019] [Indexed: 12/17/2022]
Abstract
Mutations in the gene encoding the microtubule severing ATPase spastin are the most frequent cause of hereditary spastic paraplegia, a genetic condition characterised by length-dependent axonal degeneration. Here, we show that HeLa cells lacking spastin and embryonic fibroblasts from a spastin knock-in mouse model become highly polarised and develop cellular protrusions. In HeLa cells, this phenotype was rescued by wild-type spastin, but not by forms unable to sever microtubules or interact with endosomal ESCRT-III proteins. Cells lacking the spastin-interacting ESCRT-III-associated proteins IST1 or CHMP1B also developed protrusions. The protrusion phenotype required protrudin, a RAB-interacting protein that interacts with spastin and localises to ER-endosome contact sites, where it promotes KIF5-dependent endosomal motility to protrusions. Consistent with this, the protrusion phenotype in cells lacking spastin also required KIF5. Lack or mutation of spastin resulted in functional consequences for receptor traffic of a pathway implicated in HSP, as Bone Morphogenetic Protein receptor distribution became polarised. Our results, therefore, identify a novel role for ESCRT-III proteins and spastin in regulating polarised membrane traffic.
Collapse
Affiliation(s)
- James W Connell
- Department of Medical Genetics and Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK
- Alzheimer's Research, Cambridge, UK
| | - Rachel J Allison
- Department of Medical Genetics and Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Catherine E Rodger
- Department of Medical Genetics and Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Guy Pearson
- Department of Medical Genetics and Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Eliska Zlamalova
- Department of Medical Genetics and Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Evan Reid
- Department of Medical Genetics and Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK.
| |
Collapse
|
102
|
Unrestrained ESCRT-III drives micronuclear catastrophe and chromosome fragmentation. Nat Cell Biol 2020; 22:856-867. [PMID: 32601372 DOI: 10.1038/s41556-020-0537-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/24/2020] [Indexed: 12/11/2022]
Abstract
The ESCRT-III membrane fission machinery maintains the integrity of the nuclear envelope. Although primary nuclei resealing takes minutes, micronuclear envelope ruptures seem to be irreversible. Instead, micronuclear ruptures result in catastrophic membrane collapse and are associated with chromosome fragmentation and chromothripsis, complex chromosome rearrangements thought to be a major driving force in cancer development. Here we use a combination of live microscopy and electron tomography, as well as computer simulations, to uncover the mechanism underlying micronuclear collapse. We show that, due to their small size, micronuclei inherently lack the capacity of primary nuclei to restrict the accumulation of CHMP7-LEMD2, a compartmentalization sensor that detects loss of nuclear integrity. This causes unrestrained ESCRT-III accumulation, which drives extensive membrane deformation, DNA damage and chromosome fragmentation. Thus, the nuclear-integrity surveillance machinery is a double-edged sword, as its sensitivity ensures rapid repair at primary nuclei while causing unrestrained activity at ruptured micronuclei, with catastrophic consequences for genome stability.
Collapse
|
103
|
von Appen A, LaJoie D, Johnson IE, Trnka MJ, Pick SM, Burlingame AL, Ullman KS, Frost A. LEM2 phase separation promotes ESCRT-mediated nuclear envelope reformation. Nature 2020; 582:115-118. [PMID: 32494070 PMCID: PMC7321842 DOI: 10.1038/s41586-020-2232-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 02/26/2020] [Indexed: 01/01/2023]
Abstract
During cell division, remodelling of the nuclear envelope enables chromosome segregation by the mitotic spindle1. The reformation of sealed nuclei requires ESCRTs (endosomal sorting complexes required for transport) and LEM2, a transmembrane ESCRT adaptor2-4. Here we show how the ability of LEM2 to condense on microtubules governs the activation of ESCRTs and coordinated spindle disassembly. The LEM motif of LEM2 binds BAF, conferring on LEM2 an affinity for chromatin5,6, while an adjacent low-complexity domain (LCD) promotes LEM2 phase separation. A proline-arginine-rich sequence within the LCD binds to microtubules and targets condensation of LEM2 to spindle microtubules that traverse the nascent nuclear envelope. Furthermore, the winged-helix domain of LEM2 activates the ESCRT-II/ESCRT-III hybrid protein CHMP7 to form co-oligomeric rings. Disruption of these events in human cells prevented the recruitment of downstream ESCRTs, compromised spindle disassembly, and led to defects in nuclear integrity and DNA damage. We propose that during nuclear reassembly LEM2 condenses into a liquid-like phase and coassembles with CHMP7 to form a macromolecular O-ring seal at the confluence between membranes, chromatin and the spindle. The properties of LEM2 described here, and the homologous architectures of related inner nuclear membrane proteins7,8, suggest that phase separation may contribute to other critical envelope functions, including interphase repair8-13 and chromatin organization14-17.
Collapse
Affiliation(s)
- Alexander von Appen
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Dollie LaJoie
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Isabel E Johnson
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Michael J Trnka
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah M Pick
- Faculty of Chemistry and Pharmacy, University of Freiburg, Freiburg, Germany
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Katharine S Ullman
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
104
|
Moser von Filseck J, Barberi L, Talledge N, Johnson IE, Frost A, Lenz M, Roux A. Anisotropic ESCRT-III architecture governs helical membrane tube formation. Nat Commun 2020; 11:1516. [PMID: 32471995 PMCID: PMC7260168 DOI: 10.1038/s41467-020-15327-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
ESCRT-III proteins assemble into ubiquitous membrane-remodeling polymers during many cellular processes. Here we describe the structure of helical membrane tubes that are scaffolded by bundled ESCRT-III filaments. Cryo-ET reveals how the shape of the helical membrane tube arises from the assembly of two distinct bundles of helical filaments that have the same helical path but bind the membrane with different interfaces. Higher-resolution cryo-EM of filaments bound to helical bicelles confirms that ESCRT-III filaments can interact with the membrane through a previously undescribed interface. Mathematical modeling demonstrates that the interface described above is key to the mechanical stability of helical membrane tubes and helps infer the rigidity of the described protein filaments. Altogether, our results suggest that the interactions between ESCRT-III filaments and the membrane could proceed through multiple interfaces, to provide assembly on membranes with various shapes, or adapt the orientation of the filaments towards the membrane during membrane remodeling.
Collapse
Affiliation(s)
| | - Luca Barberi
- LPTMS, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay, France.
- Biochemistry Department, University of Geneva, 1211, Geneva, Switzerland.
| | - Nathaniel Talledge
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- California Institute for Quantitative Biosciences, San Francisco, CA, 94158, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, 841112, USA
- Institute for Molecular Virology, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Isabel E Johnson
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- California Institute for Quantitative Biosciences, San Francisco, CA, 94158, USA
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94158, USA
- California Institute for Quantitative Biosciences, San Francisco, CA, 94158, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, 841112, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| | - Martin Lenz
- LPTMS, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay, France
- Laboratoire de Physique et Mécanique des Milieux Hétérogènes, UMR 7636, CNRS, ESPCI Paris, PSL Research University, Université Paris Diderot, Sorbonne Université, 75005, Paris, France
| | - Aurélien Roux
- Biochemistry Department, University of Geneva, 1211, Geneva, Switzerland.
- Swiss National Centre for Competence in Research Programme Chemical Biology, Geneva, 1211, Switzerland.
| |
Collapse
|
105
|
Bertin A, de Franceschi N, de la Mora E, Maity S, Alqabandi M, Miguet N, di Cicco A, Roos WH, Mangenot S, Weissenhorn W, Bassereau P. Human ESCRT-III polymers assemble on positively curved membranes and induce helical membrane tube formation. Nat Commun 2020; 11:2663. [PMID: 32471988 PMCID: PMC7260177 DOI: 10.1038/s41467-020-16368-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Endosomal sorting complexes for transport-III (ESCRT-III) assemble in vivo onto membranes with negative Gaussian curvature. How membrane shape influences ESCRT-III polymerization and how ESCRT-III shapes membranes is yet unclear. Human core ESCRT-III proteins, CHMP4B, CHMP2A, CHMP2B and CHMP3 are used to address this issue in vitro by combining membrane nanotube pulling experiments, cryo-electron tomography and AFM. We show that CHMP4B filaments preferentially bind to flat membranes or to tubes with positive mean curvature. Both CHMP2B and CHMP2A/CHMP3 assemble on positively curved membrane tubes. Combinations of CHMP4B/CHMP2B and CHMP4B/CHMP2A/CHMP3 are recruited to the neck of pulled membrane tubes and reshape vesicles into helical "corkscrew-like" membrane tubes. Sub-tomogram averaging reveals that the ESCRT-III filaments assemble parallel and locally perpendicular to the tube axis, highlighting the mechanical stresses imposed by ESCRT-III. Our results underline the versatile membrane remodeling activity of ESCRT-III that may be a general feature required for cellular membrane remodeling processes.
Collapse
Affiliation(s)
- Aurélie Bertin
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France.
- Sorbonne Université, 75005, Paris, France.
| | - Nicola de Franceschi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France.
- Sorbonne Université, 75005, Paris, France.
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 71, avenue des Martyrs, 38000, Grenoble, France.
| | - Eugenio de la Mora
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France
- Sorbonne Université, 75005, Paris, France
| | - Sourav Maity
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Nijenborgh 4, 9747, AG Groningen, The Netherlands
| | - Maryam Alqabandi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France
- Sorbonne Université, 75005, Paris, France
| | - Nolwen Miguet
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 71, avenue des Martyrs, 38000, Grenoble, France
| | - Aurélie di Cicco
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France
- Sorbonne Université, 75005, Paris, France
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Nijenborgh 4, 9747, AG Groningen, The Netherlands
| | - Stéphanie Mangenot
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France
- Sorbonne Université, 75005, Paris, France
| | - Winfried Weissenhorn
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 71, avenue des Martyrs, 38000, Grenoble, France.
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005, Paris, France.
- Sorbonne Université, 75005, Paris, France.
| |
Collapse
|
106
|
Flower TG, Takahashi Y, Hudait A, Rose K, Tjahjono N, Pak AJ, Yokom AL, Liang X, Wang HG, Bouamr F, Voth GA, Hurley JH. A helical assembly of human ESCRT-I scaffolds reverse-topology membrane scission. Nat Struct Mol Biol 2020; 27:570-580. [PMID: 32424346 PMCID: PMC7339825 DOI: 10.1038/s41594-020-0426-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/31/2020] [Indexed: 12/26/2022]
Abstract
The ESCRT complexes drive membrane scission in HIV-1 release, autophagosome closure, MVB biogenesis, cytokinesis, and other cell processes. ESCRT-I is the most upstream complex and bridges the system to HIV-1 Gag in virus release. The crystal structure of the headpiece of human ESCRT-I comprising TSG101–VPS28–VPS37B–MVB12A was determined, revealing an ESCRT-I helical assembly with a 12 molecule repeat. Electron microscopy confirmed that ESCRT-I subcomplexes form helical filaments in solution. Mutation of VPS28 helical interface residues blocks filament formation in vitro and autophagosome closure and HIV-1 release in human cells. Coarse grained simulations of ESCRT assembly at HIV-1 budding sites suggest that formation of a 12-membered ring of ESCRT-I molecules is a geometry-dependent checkpoint during late stages of Gag assembly and HIV-1 budding, and templates ESCRT-III assembly for membrane scission. These data show that ESCRT-I is not merely a bridging adaptor, but has an essential scaffolding and mechanical role in its own right. Further information on experimental design is available in the Nature Research Reporting Summary linked to this article.
Collapse
Affiliation(s)
- Thomas G Flower
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Yoshinori Takahashi
- Department of Pediatrics, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Arpa Hudait
- Department of Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Kevin Rose
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Tjahjono
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Alexander J Pak
- Department of Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Adam L Yokom
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Xinwen Liang
- Department of Pediatrics, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Hong-Gang Wang
- Department of Pediatrics, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Fadila Bouamr
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gregory A Voth
- Department of Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - James H Hurley
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA. .,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
107
|
Butt BG, Owen DJ, Jeffries CM, Ivanova L, Hill CH, Houghton JW, Ahmed MF, Antrobus R, Svergun DI, Welch JJ, Crump CM, Graham SC. Insights into herpesvirus assembly from the structure of the pUL7:pUL51 complex. eLife 2020; 9:e53789. [PMID: 32391791 PMCID: PMC7289601 DOI: 10.7554/elife.53789] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/07/2020] [Indexed: 12/19/2022] Open
Abstract
Herpesviruses acquire their membrane envelopes in the cytoplasm of infected cells via a molecular mechanism that remains unclear. Herpes simplex virus (HSV)-1 proteins pUL7 and pUL51 form a complex required for efficient virus envelopment. We show that interaction between homologues of pUL7 and pUL51 is conserved across human herpesviruses, as is their association with trans-Golgi membranes. We characterized the HSV-1 pUL7:pUL51 complex by solution scattering and chemical crosslinking, revealing a 1:2 complex that can form higher-order oligomers in solution, and we solved the crystal structure of the core pUL7:pUL51 heterodimer. While pUL7 adopts a previously-unseen compact fold, the helix-turn-helix conformation of pUL51 resembles the cellular endosomal complex required for transport (ESCRT)-III component CHMP4B and pUL51 forms ESCRT-III-like filaments, suggesting a direct role for pUL51 in promoting membrane scission during virus assembly. Our results provide a structural framework for understanding the role of the conserved pUL7:pUL51 complex in herpesvirus assembly.
Collapse
Affiliation(s)
- Benjamin G Butt
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Danielle J Owen
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Cy M Jeffries
- European Molecular Biology Laboratory (EMBL) Hamburg SiteHamburgGermany
| | - Lyudmila Ivanova
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Chris H Hill
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Jack W Houghton
- Cambridge Institute for Medical Research, University of CambridgeCambridgeUnited Kingdom
| | - Md Firoz Ahmed
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of CambridgeCambridgeUnited Kingdom
| | - Dmitri I Svergun
- European Molecular Biology Laboratory (EMBL) Hamburg SiteHamburgGermany
| | - John J Welch
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
| | - Colin M Crump
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Stephen C Graham
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
108
|
Sewduth R, Pandolfi S, Steklov M, Sheryazdanova A, Zhao P, Criem N, Baietti M, Lechat B, Quarck R, Impens F, Sablina A. The Noonan Syndrome Gene Lztr1 Controls Cardiovascular Function by Regulating Vesicular Trafficking. Circ Res 2020; 126:1379-1393. [PMID: 32175818 PMCID: PMC8575076 DOI: 10.1161/circresaha.119.315730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Noonan syndrome (NS) is one of the most frequent genetic disorders. Bleeding problems are among the most common, yet poorly defined complications associated with NS. A lack of consensus on the management of bleeding complications in patients with NS indicates an urgent need for new therapeutic approaches. OBJECTIVE Bleeding disorders have recently been described in patients with NS harboring mutations of LZTR1 (leucine zipper-like transcription regulator 1), an adaptor for CUL3 (CULLIN3) ubiquitin ligase complex. Here, we assessed the pathobiology of LZTR1-mediated bleeding disorders. METHODS AND RESULTS Whole-body and vascular specific knockout of Lztr1 results in perinatal lethality due to cardiovascular dysfunction. Lztr1 deletion in blood vessels of adult mice leads to abnormal vascular leakage. We found that defective adherent and tight junctions in Lztr1-depleted endothelial cells are caused by dysregulation of vesicular trafficking. LZTR1 affects the dynamics of fusion and fission of recycling endosomes by controlling ubiquitination of the ESCRT-III (endosomal sorting complex required for transport III) component CHMP1B (charged multivesicular protein 1B), whereas NS-associated LZTR1 mutations diminish CHMP1B ubiquitination. LZTR1-mediated dysregulation of CHMP1B ubiquitination triggers endosomal accumulation and subsequent activation of VEGFR2 (vascular endothelial growth factor receptor 2) and decreases blood levels of soluble VEGFR2 in Lztr1 haploinsufficient mice. Inhibition of VEGFR2 activity by cediranib rescues vascular abnormalities observed in Lztr1 knockout mice Conclusions: Lztr1 deletion phenotypically overlaps with bleeding diathesis observed in patients with NS. ELISA screening of soluble VEGFR2 in the blood of LZTR1-mutated patients with NS may predict both the severity of NS phenotypes and potential responders to anti-VEGF therapy. VEGFR inhibitors could be beneficial for the treatment of bleeding disorders in patients with NS.
Collapse
Affiliation(s)
- R. Sewduth
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - S. Pandolfi
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - M. Steklov
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - A. Sheryazdanova
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - P. Zhao
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - N. Criem
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - M.F. Baietti
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - B. Lechat
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - R. Quarck
- University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - F. Impens
- Department of Biomolecular Medicine, Ghent University, B-9000 Ghent, Belgium
- VIB Center for Medical Biotechnology, B-9000 Ghent, Belgium
- VIB Proteomics Core, Albert Baertsoenkaai 3, 9000 Ghent, Belgium
| | - A.A. Sablina
- VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
109
|
Nguyen HC, Talledge N, McCullough J, Sharma A, Moss FR, Iwasa JH, Vershinin MD, Sundquist WI, Frost A. Membrane constriction and thinning by sequential ESCRT-III polymerization. Nat Struct Mol Biol 2020; 27:392-399. [PMID: 32251413 PMCID: PMC7343221 DOI: 10.1038/s41594-020-0404-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/05/2020] [Indexed: 01/04/2023]
Abstract
The endosomal sorting complexes required for transport (ESCRTs) mediate diverse membrane remodeling events. These typically require ESCRT-III proteins to stabilize negatively curved membranes; however, recent work has indicated that certain ESCRT-IIIs also participate in positive-curvature membrane-shaping reactions. ESCRT-IIIs polymerize into membrane-binding filaments, but the structural basis for negative versus positive membrane remodeling by these proteins remains poorly understood. To learn how certain ESCRT-IIIs shape positively curved membranes, we determined structures of human membrane-bound CHMP1B-only, membrane-bound CHMP1B + IST1, and IST1-only filaments by cryo-EM. Our structures show how CHMP1B first polymerizes into a single-stranded helical filament, shaping membranes into moderate-curvature tubules. Subsequently, IST1 assembles a second strand on CHMP1B, further constricting the membrane tube and reducing its diameter nearly to the fission point. Each step of constriction thins the underlying bilayer, lowering the barrier to membrane fission. Our structures reveal how a two-component, sequential polymerization mechanism drives membrane tubulation, constriction and bilayer thinning.
Collapse
Affiliation(s)
- Henry C Nguyen
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Nathaniel Talledge
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Institute for Molecular Virology, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - John McCullough
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Abhimanyu Sharma
- Department of Physics & Astronomy, University of Utah, Salt Lake City, UT, USA
| | - Frank R Moss
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Janet H Iwasa
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Michael D Vershinin
- Department of Physics & Astronomy, University of Utah, Salt Lake City, UT, USA
- Department of Biology, University of Utah, Salt Lake City, UT, USA
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT, USA
| | - Wesley I Sundquist
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| | - Adam Frost
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
110
|
Mandal T, Lough W, Spagnolie SE, Audhya A, Cui Q. Molecular Simulation of Mechanical Properties and Membrane Activities of the ESCRT-III Complexes. Biophys J 2020; 118:1333-1343. [PMID: 32078797 PMCID: PMC7091516 DOI: 10.1016/j.bpj.2020.01.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 01/16/2023] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) machinery carries out the membrane scission reactions that are required for many biological processes throughout cells. How ESCRTs bind and deform cellular membranes and ultimately produce vesicles has been a matter of active research in recent years. In this study, we use fully atomistic molecular dynamics simulations to scrutinize the structural details of a filament composed of Vps32 protomers, a major component of ESCRT-III complexes. The simulations show that both hydrophobic and electrostatic interactions between monomers help maintain the structural stability of the filament, which exhibits an intrinsic bend and twist. Our findings suggest that the accumulation of bending and twisting stresses as the filament elongates on the membrane surface likely contributes to the driving force for membrane invagination. The filament exposes a large cationic surface that senses the negatively charged lipids in the membrane, and the N-terminal amphipathic helix of the monomers not only acts as a membrane anchor but also generates significant positive membrane curvature. Taking all results together, we discuss a plausible mechanism for membrane invagination driven by ESCRT-III.
Collapse
Affiliation(s)
- Taraknath Mandal
- Department of Chemistry, Boston University, Boston, Massachusetts
| | | | | | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Qiang Cui
- Departments of Chemistry, Physics, and Biomedical Engineering, Boston University, Boston, Massachusetts.
| |
Collapse
|
111
|
Petsalaki E, Zachos G. DNA damage response proteins regulating mitotic cell division: double agents preserving genome stability. FEBS J 2020; 287:1700-1721. [PMID: 32027459 DOI: 10.1111/febs.15240] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/10/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022]
Abstract
The DNA damage response recognizes DNA lesions and coordinates a cell cycle arrest with the repair of the damaged DNA, or removal of the affected cells to prevent the passage of genetic alterations to the next generation. The mitotic cell division, on the other hand, is a series of processes that aims to accurately segregate the genomic material from the maternal to the two daughter cells. Despite their great importance in safeguarding genomic integrity, the DNA damage response and the mitotic cell division were long viewed as unrelated processes, mainly because animal cells that are irradiated during mitosis continue cell division without repairing the broken chromosomes. However, recent studies have demonstrated that DNA damage proteins play an important role in mitotic cell division. This is performed through regulation of the onset of mitosis, mitotic spindle formation, correction of misattached kinetochore-microtubules, spindle checkpoint signaling, or completion of cytokinesis (abscission), in the absence of DNA damage. In this review, we summarize the roles of DNA damage proteins in unperturbed mitosis, analyze the molecular mechanisms involved, and discuss the potential implications of these findings in cancer therapy.
Collapse
Affiliation(s)
- Eleni Petsalaki
- Department of Biology, University of Crete, Heraklion, Greece
| | - George Zachos
- Department of Biology, University of Crete, Heraklion, Greece
| |
Collapse
|
112
|
The ESCRT-II Subunit EAP20/VPS25 and the Bro1 Domain Proteins HD-PTP and BROX Are Individually Dispensable for Herpes Simplex Virus 1 Replication. J Virol 2020; 94:JVI.01641-19. [PMID: 31748394 DOI: 10.1128/jvi.01641-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Capsid envelopment during assembly of the neurotropic herpesviruses herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV) in the infected cell cytoplasm is thought to involve the late-acting cellular ESCRT (endosomal sorting complex required for transport) components ESCRT-III and VPS4 (vacuolar protein sorting 4). However, HSV-1, unlike members of many other families of enveloped viruses, does not appear to require the ESCRT-I subunit TSG101 or the Bro1 domain-containing protein ALIX (Alg-2-interacting protein X) to recruit and activate ESCRT-III. Alternative cellular factors that are known to be capable of regulating ESCRT-III function include the ESCRT-II complex and other members of the Bro1 family. We therefore used small interfering RNA (siRNA) to knock down the essential ESCRT-II subunit EAP20/VPS25 (ELL-associated protein 20/vacuolar protein sorting 25) and the Bro1 proteins HD-PTP (His domain-containing protein tyrosine phosphatase) and BROX (Bro1 domain and CAAX motif containing). We demonstrated reductions in levels of the targeted proteins by Western blotting and used quantitative microscopic assays to confirm loss of ESCRT-II and HD-PTP function. We found that in single-step replication experiments, the final yields of HSV-1 were unchanged following loss of EAP20, HD-PTP, or BROX.IMPORTANCE HSV-1 is a pathogen of the human nervous system that uses its own virus-encoded proteins and the normal cellular ESCRT machinery to drive the construction of its envelope. How HSV-1 structural proteins interact with ESCRT components and which subsets of cellular ESCRT proteins are utilized by the virus remain largely unknown. Here, we demonstrate that an essential component of the ESCRT-II complex and two ESCRT-associated Bro1 proteins are dispensable for HSV-1 replication.
Collapse
|
113
|
Shen Q, Grome MW, Yang Y, Lin C. Engineering Lipid Membranes with Programmable DNA Nanostructures. ADVANCED BIOSYSTEMS 2020; 4:1900215. [PMID: 31934608 PMCID: PMC6957268 DOI: 10.1002/adbi.201900215] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Indexed: 12/18/2022]
Abstract
Lipid and DNA are abundant biomolecules with critical functions in cells. The water-insoluble, amphipathic lipid molecules are best known for their roles in energy storage (e.g. as triglyceride), signaling (e.g. as sphingolipid), and compartmentalization (e.g. by forming membrane-enclosed bodies). The soluble, highly negatively charged DNA, which stores cells' genetic information, has proven to be an excellent material for constructing programmable nanostructures in vitro thanks to its self-assembling capabilities. These two seemingly distant molecules make contact within cell nuclei, often via lipidated proteins, with proposed functions of modulating chromatin structures. Carefully formulated lipid/DNA complexes are promising reagents for gene therapy. The past few years saw an emerging research field of interfacing DNA nanostructures with lipid membranes, with an overarching goal of generating DNA/lipid hybrid materials that possess novel and controllable structure, dynamics, and function. An arsenal of DNA-based tools has been created to coat, mold, deform, and penetrate lipid bilayers, affording us the ability to manipulate membranes with nanoscopic precision. These membrane engineering methods not only enable quantitative biophysical studies, but also open new opportunities in synthetic biology (e.g. artificial cells) and therapeutics (e.g. drug delivery).
Collapse
Affiliation(s)
- Qi Shen
- Department of Cell Biology and Nanobiology Institute, Yale University
| | - Michael W Grome
- Department of Cell Biology and Nanobiology Institute, Yale University
| | - Yang Yang
- Department of Cell Biology and Nanobiology Institute, Yale University
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Chenxiang Lin
- Department of Cell Biology and Nanobiology Institute, Yale University
| |
Collapse
|
114
|
Catalano M, O'Driscoll L. Inhibiting extracellular vesicles formation and release: a review of EV inhibitors. J Extracell Vesicles 2019; 9:1703244. [PMID: 32002167 PMCID: PMC6968539 DOI: 10.1080/20013078.2019.1703244] [Citation(s) in RCA: 438] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 11/14/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
It is now becoming well established that vesicles are released from a broad range of cell types and are involved in cell-to-cell communication, both in physiological and pathological conditions. Once outside the cell, these vesicles are termed extracellular vesicles (EVs). The cellular origin (cell type), subcellular origin (through the endosomal pathway or pinched from the cell membrane) and content (what proteins, glycoproteins, lipids, nucleic acids, metabolites) are transported by the EVs, and their size, all seem to be contributing factors to their overall heterogeneity. Efforts are being invested into attempting to block the release of subpopulations of EVs or, indeed, all EVs. Some such studies are focussed on investigating EV inhibitors as research tools; others are interested in the longerterm potential of using such inhibitors in pathological conditions such as cancer. This review, intended to be of relevance to both researchers already well established in the EV field and newcomers to this field, provides an outline of the compounds that have been most extensively studied for this purpose, their proposed mechanisms of actions and the findings of these studies.
Collapse
Affiliation(s)
- Mariadelva Catalano
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
115
|
Diwaker D, Wilson DW. Microtubule-Dependent Trafficking of Alphaherpesviruses in the Nervous System: The Ins and Outs. Viruses 2019; 11:v11121165. [PMID: 31861082 PMCID: PMC6950448 DOI: 10.3390/v11121165] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 12/12/2022] Open
Abstract
The Alphaherpesvirinae include the neurotropic pathogens herpes simplex virus and varicella zoster virus of humans and pseudorabies virus of swine. These viruses establish lifelong latency in the nuclei of peripheral ganglia, but utilize the peripheral tissues those neurons innervate for productive replication, spread, and transmission. Delivery of virions from replicative pools to the sites of latency requires microtubule-directed retrograde axonal transport from the nerve terminus to the cell body of the sensory neuron. As a corollary, during reactivation newly assembled virions must travel along axonal microtubules in the anterograde direction to return to the nerve terminus and infect peripheral tissues, completing the cycle. Neurotropic alphaherpesviruses can therefore exploit neuronal microtubules and motors for long distance axonal transport, and alternate between periods of sustained plus end- and minus end-directed motion at different stages of their infectious cycle. This review summarizes our current understanding of the molecular details by which this is achieved.
Collapse
Affiliation(s)
- Drishya Diwaker
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Duncan W. Wilson
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
- Correspondence: ; Tel.: +1-(718)-430-2305
| |
Collapse
|
116
|
Abstract
Cellular membranes can form two principally different involutions, which either exclude or contain cytosol. The 'classical' budding reactions, such as those occurring during endocytosis or formation of exocytic vesicles, involve proteins that assemble on the cytosol-excluding face of the bud neck. Inverse membrane involution occurs in a wide range of cellular processes, supporting cytokinesis, endosome maturation, autophagy, membrane repair and many other processes. Such inverse membrane remodelling is mediated by a heteromultimeric protein machinery known as endosomal sorting complex required for transport (ESCRT). ESCRT proteins assemble on the cytosolic (or nucleoplasmic) face of the neck of the forming involution and cooperate with the ATPase VPS4 to drive membrane scission or sealing. Here, we review similarities and differences of various ESCRT-dependent processes, with special emphasis on mechanisms of ESCRT recruitment.
Collapse
|
117
|
Petsalaki E, Zachos G. Building bridges between chromosomes: novel insights into the abscission checkpoint. Cell Mol Life Sci 2019; 76:4291-4307. [PMID: 31302750 PMCID: PMC11105294 DOI: 10.1007/s00018-019-03224-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/19/2019] [Accepted: 07/05/2019] [Indexed: 12/20/2022]
Abstract
In the presence of chromatin bridges, mammalian cells delay completion of cytokinesis (abscission) to prevent chromatin breakage or tetraploidization by regression of the cleavage furrow. This abscission delay is called "the abscission checkpoint" and is dependent on Aurora B kinase. Furthermore, cells stabilize the narrow cytoplasmic canal between the two daughter cells until the DNA bridges are resolved. Impaired abscission checkpoint signaling or unstable intercellular canals can lead to accumulation of DNA damage, aneuploidy, or generation of polyploid cells which are associated with tumourigenesis. However, the molecular mechanisms involved have only recently started to emerge. In this review, we focus on the molecular pathways of the abscission checkpoint and describe newly identified triggers, Aurora B-regulators and effector proteins in abscission checkpoint signaling. We also describe mechanisms that control intercellular bridge stabilization, DNA bridge resolution, or abscission checkpoint silencing upon satisfaction, and discuss how abscission checkpoint proteins can be targeted to potentially improve cancer therapy.
Collapse
Affiliation(s)
- Eleni Petsalaki
- Department of Biology, University of Crete, Vassilika Vouton, 70013, Heraklion, Greece
| | - George Zachos
- Department of Biology, University of Crete, Vassilika Vouton, 70013, Heraklion, Greece.
| |
Collapse
|
118
|
Ladinsky MS, Khamaikawin W, Jung Y, Lin S, Lam J, An DS, Bjorkman PJ, Kieffer C. Mechanisms of virus dissemination in bone marrow of HIV-1-infected humanized BLT mice. eLife 2019; 8:46916. [PMID: 31657719 PMCID: PMC6839903 DOI: 10.7554/elife.46916] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 10/27/2019] [Indexed: 02/06/2023] Open
Abstract
Immune progenitor cells differentiate in bone marrow (BM) and then migrate to tissues. HIV-1 infects multiple BM cell types, but virus dissemination within BM has been poorly understood. We used light microscopy and electron tomography to elucidate mechanisms of HIV-1 dissemination within BM of HIV-1–infected BM/liver/thymus (BLT) mice. Tissue clearing combined with confocal and light sheet fluorescence microscopy revealed distinct populations of HIV-1 p24-producing cells in BM early after infection, and quantification of these populations identified macrophages as the principal subset of virus-producing cells in BM over time. Electron tomography demonstrated three modes of HIV-1 dissemination in BM: (i) semi-synchronous budding from T-cell and macrophage membranes, (ii) mature virus association with virus-producing T-cell uropods contacting putative target cells, and (iii) macrophages engulfing HIV-1–producing T-cells and producing virus within enclosed intracellular compartments that fused to invaginations with access to the extracellular space. These results illustrate mechanisms by which the specialized environment of the BM can promote virus spread locally and to distant lymphoid tissues.
Collapse
Affiliation(s)
- Mark S Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Wannisa Khamaikawin
- School of Nursing, UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yujin Jung
- School of Nursing, UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, United States
| | - Samantha Lin
- School of Nursing, UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, United States
| | - Jennifer Lam
- School of Nursing, UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, United States
| | - Dong Sung An
- School of Nursing, UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, United States
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Collin Kieffer
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
119
|
Harker-Kirschneck L, Baum B, Šarić A. Changes in ESCRT-III filament geometry drive membrane remodelling and fission in silico. BMC Biol 2019; 17:82. [PMID: 31640700 PMCID: PMC6806514 DOI: 10.1186/s12915-019-0700-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/17/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND ESCRT-III is a membrane remodelling filament with the unique ability to cut membranes from the inside of the membrane neck. It is essential for the final stage of cell division, the formation of vesicles, the release of viruses, and membrane repair. Distinct from other cytoskeletal filaments, ESCRT-III filaments do not consume energy themselves, but work in conjunction with another ATP-consuming complex. Despite rapid progress in describing the cell biology of ESCRT-III, we lack an understanding of the physical mechanisms behind its force production and membrane remodelling. RESULTS Here we present a minimal coarse-grained model that captures all the experimentally reported cases of ESCRT-III driven membrane sculpting, including the formation of downward and upward cones and tubules. This model suggests that a change in the geometry of membrane bound ESCRT-III filaments-from a flat spiral to a 3D helix-drives membrane deformation. We then show that such repetitive filament geometry transitions can induce the fission of cargo-containing vesicles. CONCLUSIONS Our model provides a general physical mechanism that explains the full range of ESCRT-III-dependent membrane remodelling and scission events observed in cells. This mechanism for filament force production is distinct from the mechanisms described for other cytoskeletal elements discovered so far. The mechanistic principles revealed here suggest new ways of manipulating ESCRT-III-driven processes in cells and could be used to guide the engineering of synthetic membrane-sculpting systems.
Collapse
Affiliation(s)
- Lena Harker-Kirschneck
- Department of Physics & Astronomy, University College London, Gower Street, London, WC1E 6BT UK
- Institute for the Physics of Living Systems, University College London, Gower Street, London, WC1E 6BT UK
| | - Buzz Baum
- Institute for the Physics of Living Systems, University College London, Gower Street, London, WC1E 6BT UK
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - And̄ela Šarić
- Department of Physics & Astronomy, University College London, Gower Street, London, WC1E 6BT UK
- Institute for the Physics of Living Systems, University College London, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
120
|
Fowler PC, Garcia-Pardo ME, Simpson JC, O'Sullivan NC. NeurodegenERation: The Central Role for ER Contacts in Neuronal Function and Axonopathy, Lessons From Hereditary Spastic Paraplegias and Related Diseases. Front Neurosci 2019; 13:1051. [PMID: 31680803 PMCID: PMC6801308 DOI: 10.3389/fnins.2019.01051] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/19/2019] [Indexed: 12/17/2022] Open
Abstract
The hereditary spastic paraplegias (HSPs) are a group of inherited neurodegenerative conditions whose characteristic feature is degeneration of the longest axons within the corticospinal tract which leads to progressive spasticity and weakness of the lower limbs. Though highly genetically heterogeneous, the majority of HSP cases are caused by mutations in genes encoding proteins that are responsible for generating and organizing the tubular endoplasmic reticulum (ER). Despite this, the role of the ER within neurons, particularly the long axons affected in HSP, is not well understood. Throughout axons, ER tubules make extensive contacts with other organelles, the cytoskeleton and the plasma membrane. At these ER contacts, protein complexes work in concert to perform specialized functions including organelle shaping, calcium homeostasis and lipid biogenesis, all of which are vital for neuronal survival and may be disrupted by HSP-causing mutations. In this article we summarize the proteins which mediate ER contacts, review the functions these contacts are known to carry out within neurons, and discuss the potential contribution of disruption of ER contacts to axonopathy in HSP.
Collapse
Affiliation(s)
- Philippa C Fowler
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - M Elena Garcia-Pardo
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Jeremy C Simpson
- UCD School of Biology and Environmental Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Niamh C O'Sullivan
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
121
|
Abstract
Lipid droplets (LDs) store energy-rich fatty acids (FAs), but to harvest them, LDs donate FAs to peroxisomes for oxidation. In this issue, Chang et al. (2019. J. Cell Biol. https://doi.org/10.1083/jcb.201902061) identify M1 Spastin as an LD-peroxisome tether that interacts with ESCRT-III components to facilitate lipid exchange between these two organelles.
Collapse
Affiliation(s)
- W. Mike Henne
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
122
|
Gatta AT, Carlton JG. The ESCRT-machinery: closing holes and expanding roles. Curr Opin Cell Biol 2019; 59:121-132. [DOI: 10.1016/j.ceb.2019.04.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 01/08/2023]
|
123
|
Romanov V, McCullough J, Gale BK, Frost A. A Tunable Microfluidic Device Enables Cargo Encapsulation by Cell- or Organelle-Sized Lipid Vesicles Comprising Asymmetric Lipid Bilayers. ADVANCED BIOSYSTEMS 2019; 3:1900010. [PMID: 31428671 PMCID: PMC6699779 DOI: 10.1002/adbi.201900010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Indexed: 01/14/2023]
Abstract
Cellular membranes play host to a wide variety of morphologically and chemically complex processes. Although model membranes, like liposomes, are already widely used to reconstitute and study these processes, better tools are needed for making model bilayers that faithfully mimic cellular membranes. Existing methods for fabricating cell-sized (μm) or organelle-sized (tens to hundreds of nanometers) lipid vesicles have distinctly different requirements. Of particular note for biology, it remains challenging for any technique to efficiently encapsulate fragile cargo molecules or to generate liposomes with stable, asymmetric lipid leaflets within the bilayer. Here a tunable microfluidic device and protocol for fabricating liposomes with desired diameters ranging from ≈10 μm to ≈100 nm are described. Lipid vesicle size is templated by the simple inclusion of a polycarbonate filter within the microfluidic system and tuned with flow rate. It is shown that the vesicles made with this device are stable, unilamellar, lipid asymmetric, and capable of supporting transmembrane protein assembly, peripheral membrane protein binding, as well as soluble cargo encapsulation (including designer nanocages for biotechnology applications). These fabricated vesicles provide a new platform for studying the biophysically rich processes found within lipid-lipid and lipid-protein systems typically associated with cellular membranes.
Collapse
Affiliation(s)
- Valentin Romanov
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - John McCullough
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Bruce K Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam Frost
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA; California Institute for Quantitative Biomedical Research, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
124
|
Feng Q, Luo Y, Zhang XN, Yang XF, Hong XY, Sun DS, Li XC, Hu Y, Li XG, Zhang JF, Li X, Yang Y, Wang Q, Liu GP, Wang JZ. MAPT/Tau accumulation represses autophagy flux by disrupting IST1-regulated ESCRT-III complex formation: a vicious cycle in Alzheimer neurodegeneration. Autophagy 2019; 16:641-658. [PMID: 31223056 DOI: 10.1080/15548627.2019.1633862] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Macroautophagy/autophagy deficit induces intracellular MAPT/tau accumulation, the hallmark pathology in Alzheimer disease (AD) and other tauopathies; however, the reverse role of MAPT accumulation in autophagy and neurodegeneration is not clear. Here, we found that overexpression of human wild-type full-length MAPT, which models MAPT pathologies as seen in sporadic AD patients, induced autophagy deficits via repression of autophagosome-lysosome fusion leading to significantly increased LC3 (microtubule-associated protein 1 light chain 3)-II and SQSTM1/p62 (sequestosome 1) protein levels with autophagosome accumulation. At the molecular level, intracellular MAPT aggregation inhibited expression of IST1 (IST1 factor associated with ESCRT-III), a positive modulator for the formation of ESCRT (the Endosomal Sorting Complex Required for Transport) complex that is required for autophagosome-lysosome fusion. Upregulating IST1 in human MAPT transgenic mice attenuated autophagy deficit with reduced MAPT aggregation and ameliorated synaptic plasticity and cognitive functions, while downregulating IST1 per se induced autophagy deficit with impaired synapse and cognitive function in naïve mice. IST1 can facilitate association of CHMP2B (charged multivesicular body protein 2B) and CHMP4B/SNF7-2 to form ESCRT-III complex, while lack of IST1 impeded the complex formation. Finally, we demonstrate that MAPT accumulation suppresses IST1 transcription with the mechanisms involving the ANP32A-regulated mask of histone acetylation. Our findings suggest that the AD-like MAPT accumulation can repress autophagosome-lysosome fusion by deregulating ANP32A-INHAT-IST1-ESCRT-III pathway, which also reveals a vicious cycle of MAPT accumulation and autophagy deficit in the chronic course of AD neurodegeneration.Abbreviations: AAV: adeno-associated virus; Aβ: β-amyloid; aCSF: artificial cerebrospinal fluid; AD: Alzheimer disease; ANP32A: acidic nuclear phosphoprotein 32 family member A; ATG: autophagy related; AVs: autophagic vacuoles; CEBPB: CCAAT enhancer binding protein beta; CHMP: charged multivesicular body protein; DMEM: Dulbecco's modified eagle's medium; EBSS: Earle's balanced salt solution; EGFR: epidermal growth factor receptor; ESCRT: endosomal sorting complex required for transport; fEPSPs: field excitatory postsynaptic potentials; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GSK3B: glycogen synthase kinase 3 beta; HAT: histone acetyl transferase; HDAC: histone deacetylase; INHAT: inhibitor of histone acetyl transferase; IST1: IST1 factor associated with ESCRT-III; LAMP2: lysosomal associated membrane protein 2; LTP: long-term potentiation; MAP1LC3: microtubule associated protein 1 light chain 3; MAPT/tau: microtubule associated protein tau; MVB: multivesicular bodies; MWM: Morris water maze; PBS: phosphate-buffered saline solution; RAB7: member RAS oncogene family; SNAREs: soluble N-ethylmaleimide-sensitive factor attachment protein receptors; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Qiong Feng
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Luo
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang-Nan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xi-Fei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiao-Yue Hong
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong-Shen Sun
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia-Chun Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Guang Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun-Fei Zhang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| |
Collapse
|
125
|
Banjade S, Tang S, Shah YH, Emr SD. Electrostatic lateral interactions drive ESCRT-III heteropolymer assembly. eLife 2019; 8:e46207. [PMID: 31246173 PMCID: PMC6663469 DOI: 10.7554/elife.46207] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Self-assembly of ESCRT-III complex is a critical step in all ESCRT-dependent events. ESCRT-III hetero-polymers adopt variable architectures, but the mechanisms of inter-subunit recognition in these hetero-polymers to create flexible architectures remain unclear. We demonstrate in vivo and in vitro that the Saccharomyces cerevisiae ESCRT-III subunit Snf7 uses a conserved acidic helix to recruit its partner Vps24. Charge-inversion mutations in this helix inhibit Snf7-Vps24 lateral interactions in the polymer, while rebalancing the charges rescues the functional defects. These data suggest that Snf7-Vps24 assembly occurs through electrostatic interactions on one surface, rather than through residue-to-residue specificity. We propose a model in which these cooperative electrostatic interactions in the polymer propagate to allow for specific inter-subunit recognition, while sliding of laterally interacting polymers enable changes in architecture at distinct stages of vesicle biogenesis. Our data suggest a mechanism by which interaction specificity and polymer flexibility can be coupled in membrane-remodeling heteropolymeric assemblies.
Collapse
Affiliation(s)
- Sudeep Banjade
- Weill Institute for Cell and Molecular BiologyCornell UniversityIthacaUnited States
- Department of Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Shaogeng Tang
- Weill Institute for Cell and Molecular BiologyCornell UniversityIthacaUnited States
- Department of Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Yousuf H Shah
- Weill Institute for Cell and Molecular BiologyCornell UniversityIthacaUnited States
- Department of Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Scott D Emr
- Weill Institute for Cell and Molecular BiologyCornell UniversityIthacaUnited States
- Department of Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| |
Collapse
|
126
|
Chang CL, Weigel AV, Ioannou MS, Pasolli HA, Xu CS, Peale DR, Shtengel G, Freeman M, Hess HF, Blackstone C, Lippincott-Schwartz J. Spastin tethers lipid droplets to peroxisomes and directs fatty acid trafficking through ESCRT-III. J Cell Biol 2019; 218:2583-2599. [PMID: 31227594 PMCID: PMC6683741 DOI: 10.1083/jcb.201902061] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/29/2019] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
Lipid droplets (LDs) are neutral lipid storage organelles that transfer lipids to various organelles including peroxisomes. Here, we show that the hereditary spastic paraplegia protein M1 Spastin, a membrane-bound AAA ATPase found on LDs, coordinates fatty acid (FA) trafficking from LDs to peroxisomes through two interrelated mechanisms. First, M1 Spastin forms a tethering complex with peroxisomal ABCD1 to promote LD-peroxisome contact formation. Second, M1 Spastin recruits the membrane-shaping ESCRT-III proteins IST1 and CHMP1B to LDs via its MIT domain to facilitate LD-to-peroxisome FA trafficking, possibly through IST1- and CHMP1B-dependent modifications in LD membrane morphology. Furthermore, LD-to-peroxisome FA trafficking mediated by M1 Spastin is required to relieve LDs of lipid peroxidation. M1 Spastin's dual roles in tethering LDs to peroxisomes and in recruiting ESCRT-III components to LD-peroxisome contact sites for FA trafficking may underlie the pathogenesis of diseases associated with defective FA metabolism in LDs and peroxisomes.
Collapse
Affiliation(s)
- Chi-Lun Chang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Aubrey V Weigel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Maria S Ioannou
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - H Amalia Pasolli
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - C Shan Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - David R Peale
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Gleb Shtengel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Melanie Freeman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Harald F Hess
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Craig Blackstone
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
127
|
Abstract
The Herpesviridae are structurally complex DNA viruses whose capsids undergo primary envelopment at the inner nuclear membrane and secondary envelopment at organelles in the cytoplasm. In both locations, there is evidence that envelope formation and scission involve the participation of multiple viral proteins and also the cellular ESCRT apparatus. It nevertheless appears that the best-understood viral strategies for ESCRT recruitment, those adopted by the retroviruses and many other families of enveloped RNA viruses, are not utilized by the Herpesviridae, at least during envelopment in the cytoplasm. Thus, although a large number of herpesvirus proteins have been assigned roles in envelopment, there is a dearth of candidates for the acquisition of the ESCRT complex and the control of envelope scission. This review summarizes our current understanding of ESCRT association by enveloped viruses, examines what is known of herpesvirus ESCRT utilization in the nucleus and cytoplasm, and identifies candidate cellular and viral proteins that could link enveloping herpesviruses to cellular ESCRT components.
Collapse
|
128
|
Abstract
Dynamin proteins assemble into characteristic helical structures around necks of clathrin-coated membrane buds. Hydrolysis of dynamin-bound GTP results in both fission of the membrane neck and partial disruption of the dynamin oligomer. Imaging by atomic force microscopy reveals that, on GTP hydrolysis, dynamin oligomers undergo a dynamic remodeling and lose their distinctive helical shape. While breakup of the dynamin helix is a critical stage in clathrin-mediated endocytosis, the mechanism for this remodeling of the oligomer has not been resolved. In this paper, we formulate an analytical, elasticity-based model for the reshaping and disassembly of the dynamin scaffold. We predict that the shape of the oligomer is modulated by the orientation of dynamin's pleckstrin homology (PH) domain relative to the underlying membrane. Our results indicate that tilt of the PH domain drives deformation and fragmentation of the oligomer, in agreement with experimental observations. This model motivated the introduction of the tilted helix: a curve that maintains a fixed angle between its normal and the normal of the embedding surface. Our findings highlight the importance of tilt as a key regulator of size and morphology of membrane-bound oligomers.
Collapse
|
129
|
Sønder SL, Boye TL, Tölle R, Dengjel J, Maeda K, Jäättelä M, Simonsen AC, Jaiswal JK, Nylandsted J. Annexin A7 is required for ESCRT III-mediated plasma membrane repair. Sci Rep 2019; 9:6726. [PMID: 31040365 PMCID: PMC6491720 DOI: 10.1038/s41598-019-43143-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/15/2019] [Indexed: 12/21/2022] Open
Abstract
The plasma membrane of eukaryotic cells forms the essential barrier to the extracellular environment, and thus plasma membrane disruptions pose a fatal threat to cells. Here, using invasive breast cancer cells we show that the Ca2+ - and phospholipid-binding protein annexin A7 is part of the plasma membrane repair response by enabling assembly of the endosomal sorting complex required for transport (ESCRT) III. Following injury to the plasma membrane and Ca2+ flux into the cytoplasm, annexin A7 forms a complex with apoptosis linked gene-2 (ALG-2) to facilitate proper recruitment and binding of ALG-2 and ALG-2-interacting protein X (ALIX) to the damaged membrane. ALG-2 and ALIX assemble the ESCRT III complex, which helps excise and shed the damaged portion of the plasma membrane during wound healing. Our results reveal a novel function of annexin A7 – enabling plasma membrane repair by regulating ESCRT III-mediated shedding of injured plasma membrane.
Collapse
Affiliation(s)
- Stine Lauritzen Sønder
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Theresa Louise Boye
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Regine Tölle
- Department of Dermatology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Dermatology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Kenji Maeda
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Marja Jäättelä
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Adam Cohen Simonsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230, Odense M, Denmark
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA
| | - Jesper Nylandsted
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark. .,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
130
|
Thaller DJ, Allegretti M, Borah S, Ronchi P, Beck M, Lusk CP. An ESCRT-LEM protein surveillance system is poised to directly monitor the nuclear envelope and nuclear transport system. eLife 2019; 8:e45284. [PMID: 30942170 PMCID: PMC6461442 DOI: 10.7554/elife.45284] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/02/2019] [Indexed: 12/22/2022] Open
Abstract
The integrity of the nuclear membranes coupled to the selective barrier of nuclear pore complexes (NPCs) are essential for the segregation of nucleoplasm and cytoplasm. Mechanical membrane disruption or perturbation to NPC assembly triggers an ESCRT-dependent surveillance system that seals nuclear pores: how these pores are sensed and sealed is ill defined. Using a budding yeast model, we show that the ESCRT Chm7 and the integral inner nuclear membrane (INM) protein Heh1 are spatially segregated by nuclear transport, with Chm7 being actively exported by Xpo1/Crm1. Thus, the exposure of the INM triggers surveillance with Heh1 locally activating Chm7. Sites of Chm7 hyperactivation show fenestrated sheets at the INM and potential membrane delivery at sites of nuclear envelope herniation. Our data suggest that perturbation to the nuclear envelope barrier would lead to local nuclear membrane remodeling to promote membrane sealing. Our findings have implications for disease mechanisms linked to NPC assembly and nuclear envelope integrity.
Collapse
Affiliation(s)
- David J Thaller
- Department of Cell BiologyYale School of MedicineNew HavenUnited States
| | - Matteo Allegretti
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryMeyerhofstrasseGermany
| | - Sapan Borah
- Department of Cell BiologyYale School of MedicineNew HavenUnited States
| | - Paolo Ronchi
- Electron Microscopy Core FacilityEuropean Molecular Biology LaboratoryMeyerhofstrasseGermany
| | - Martin Beck
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryMeyerhofstrasseGermany
| | - C Patrick Lusk
- Department of Cell BiologyYale School of MedicineNew HavenUnited States
| |
Collapse
|
131
|
Maity S, Caillat C, Miguet N, Sulbaran G, Effantin G, Schoehn G, Roos WH, Weissenhorn W. VPS4 triggers constriction and cleavage of ESCRT-III helical filaments. SCIENCE ADVANCES 2019; 5:eaau7198. [PMID: 30989108 PMCID: PMC6457934 DOI: 10.1126/sciadv.aau7198] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 02/21/2019] [Indexed: 05/19/2023]
Abstract
Many cellular processes such as endosomal vesicle budding, virus budding, and cytokinesis require extensive membrane remodeling by the endosomal sorting complex required for transport III (ESCRT-III). ESCRT-III protein family members form spirals with variable diameters in vitro and in vivo inside tubular membrane structures, which need to be constricted to proceed to membrane fission. Here, we show, using high-speed atomic force microscopy and electron microscopy, that the AAA-type adenosine triphosphatase VPS4 constricts and cleaves ESCRT-III CHMP2A-CHMP3 helical filaments in vitro. Constriction starts asymmetrically and progressively decreases the diameter of CHMP2A-CHMP3 tubular structure, thereby coiling up the CHMP2A-CHMP3 filaments into dome-like end caps. Our results demonstrate that VPS4 actively constricts ESCRT-III filaments and cleaves them before their complete disassembly. We propose that the formation of ESCRT-III dome-like end caps by VPS4 within a membrane neck structure constricts the membrane to set the stage for membrane fission.
Collapse
Affiliation(s)
- Sourav Maity
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Nijenborgh 4, 9747 AG Groningen, Netherlands
| | - Christophe Caillat
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Nolwenn Miguet
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Guidenn Sulbaran
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Gregory Effantin
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Guy Schoehn
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Wouter H. Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Nijenborgh 4, 9747 AG Groningen, Netherlands
- Corresponding author. (W.H.R.); (W.W.)
| | - Winfried Weissenhorn
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
- Corresponding author. (W.H.R.); (W.W.)
| |
Collapse
|
132
|
The role of VPS4 in ESCRT-III polymer remodeling. Biochem Soc Trans 2019; 47:441-448. [DOI: 10.1042/bst20180026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 01/04/2023]
Abstract
Abstract
The endosomal sorting complex required for transport-III (ESCRT-III) and VPS4 catalyze a variety of membrane-remodeling processes in eukaryotes and archaea. Common to these processes is the dynamic recruitment of ESCRT-III proteins from the cytosol to the inner face of a membrane neck structure, their activation and filament formation inside or at the membrane neck and the subsequent or concomitant recruitment of the AAA-type ATPase VPS4. The dynamic assembly of ESCRT-III filaments and VPS4 on cellular membranes induces constriction of membrane necks with large diameters such as the cytokinetic midbody and necks with small diameters such as those of intraluminal vesicles or enveloped viruses. The two processes seem to use different sets of ESCRT-III filaments. Constriction is then thought to set the stage for membrane fission. Here, we review recent progress in understanding the structural transitions of ESCRT-III proteins required for filament formation, the functional role of VPS4 in dynamic ESCRT-III assembly and its active role in filament constriction. The recent data will be discussed in the context of different mechanistic models for inside-out membrane fission.
Collapse
|
133
|
Schöneberg J, Pavlin MR, Yan S, Righini M, Lee IH, Carlson LA, Bahrami AH, Goldman DH, Ren X, Hummer G, Bustamante C, Hurley JH. ATP-dependent force generation and membrane scission by ESCRT-III and Vps4. Science 2019; 362:1423-1428. [PMID: 30573630 DOI: 10.1126/science.aat1839] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 09/17/2018] [Accepted: 11/07/2018] [Indexed: 12/23/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRTs) catalyze reverse-topology scission from the inner face of membrane necks in HIV budding, multivesicular endosome biogenesis, cytokinesis, and other pathways. We encapsulated ESCRT-III subunits Snf7, Vps24, and Vps2 and the AAA+ ATPase (adenosine triphosphatase) Vps4 in giant vesicles from which membrane nanotubes reflecting the correct topology of scission could be pulled. Upon ATP release by photo-uncaging, this system generated forces within the nanotubes that led to membrane scission in a manner dependent upon Vps4 catalytic activity and Vps4 coupling to the ESCRT-III proteins. Imaging of scission revealed Snf7 and Vps4 puncta within nanotubes whose presence followed ATP release, correlated with force generation and nanotube constriction, and preceded scission. These observations directly verify long-standing predictions that ATP-hydrolyzing assemblies of ESCRT-III and Vps4 sever membranes.
Collapse
Affiliation(s)
- Johannes Schöneberg
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Mark Remec Pavlin
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shannon Yan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Maurizio Righini
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Il-Hyung Lee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lars-Anders Carlson
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Amir Houshang Bahrami
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Daniel H Goldman
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xuefeng Ren
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany.,Institute of Biophysics, Goethe University, Frankfurt/M, Germany
| | - Carlos Bustamante
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA. .,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA 94720, USA.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - James H Hurley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA. .,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA 94720, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| |
Collapse
|
134
|
Structure and mechanism of the ESCRT pathway AAA+ ATPase Vps4. Biochem Soc Trans 2019; 47:37-45. [PMID: 30647138 PMCID: PMC6393862 DOI: 10.1042/bst20180260] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 01/05/2023]
Abstract
The progression of ESCRT (Endosomal Sorting Complexes Required for Transport) pathways, which mediate numerous cellular membrane fission events, is driven by the enzyme Vps4. Understanding of Vps4 mechanism is, therefore, of fundamental importance in its own right and, moreover, it is highly relevant to the understanding of many related AAA+ ATPases that function in multiple facets of cell biology. Vps4 unfolds its ESCRT-III protein substrates by translocating them through its central hexameric pore, thereby driving membrane fission and recycling of ESCRT-III subunits. This mini-review focuses on recent advances in Vps4 structure and mechanism, including ideas about how Vps4 translocates and unfolds ESCRT-III subunits. Related AAA+ ATPases that share structural features with Vps4 and likely utilize an equivalent mechanism are also discussed.
Collapse
|
135
|
Abstract
Extracellular vesicles (EVs), and exosomes in particular, were initially considered as "garbage bags" for secretion of undesired cellular components. This view has changed considerably over the last two decades, and exosomes have now emerged as important organelles controlling cell-to-cell signaling. They are present in biological fluids and have important roles in the communication between cells in physiological and pathological processes. They are envisioned for clinical use as carriers of biomarkers, therapeutic targets, and vehicles for drug delivery. Important efforts are being made to characterize the contents of these vesicles and to understand the mechanisms that govern their biogenesis and modes of action. This chapter aims to recapitulate the place given to lipids in our understanding of exosome biology. Besides their structural role and their function as carriers, certain lipids and lipid-modifying enzymes seem to exert privileged functions in this mode of cellular communication. By extension, the use of selective "lipid inhibitors" might turn out to be interesting modulators of exosomal-based cell signaling.
Collapse
Affiliation(s)
- Antonio Luis Egea-Jimenez
- Centre de Recherche en Cancérologie de Marseille, Equipe labellisée Ligue 2018, Aix-Marseille Université, Inserm, CNRS, Institut Paoli Calmettes, Marseille, France.,Department of Human Genetics, K. U. Leuven, Leuven, Belgium
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille, Equipe labellisée Ligue 2018, Aix-Marseille Université, Inserm, CNRS, Institut Paoli Calmettes, Marseille, France. .,Department of Human Genetics, K. U. Leuven, Leuven, Belgium.
| |
Collapse
|
136
|
Haag C, Klein T, Feldbrügge M. ESCRT Mutant Analysis and Imaging of ESCRT Components in the Model Fungus Ustilago maydis. Methods Mol Biol 2019; 1998:251-271. [PMID: 31250308 DOI: 10.1007/978-1-4939-9492-2_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The ESCRT machinery (endosomal sorting complex required for transport) is an evolutionarily highly conserved multiprotein complex involved in numerous cellular processes like endocytosis, membrane repair, or endosomal long-distance transport. In fungal hyphae, endocytosis and long-distance mRNA transport are tightly linked, as endocytotic vesicles are also the key carrier vehicles for mRNAs. Studying the regulatory component Did2 (CHMP1) in the plant pathogen Ustilago maydis revealed that loss of Did2 resulted in disturbed endosomal maturation, thereby causing defects in microtubule-dependent transport of early endosomes. Here, we describe methods and protocols that allow studying the role of ESCRT components during endosomal transport. We present experimental strategies to analyze U. maydis ESCRT mutant phenotypes and test complementation with heterologous components, such as ESCRT regulators from Drosophila melanogaster.
Collapse
Affiliation(s)
- Carl Haag
- Cluster of Excellence on Plant Sciences (CEPLAS), Institute for Microbiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Feldbrügge
- Cluster of Excellence on Plant Sciences (CEPLAS), Institute for Microbiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
137
|
Farré JC, Mahalingam SS, Proietto M, Subramani S. Peroxisome biogenesis, membrane contact sites, and quality control. EMBO Rep 2018; 20:embr.201846864. [PMID: 30530632 DOI: 10.15252/embr.201846864] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/08/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022] Open
Abstract
Peroxisomes are conserved organelles of eukaryotic cells with important roles in cellular metabolism, human health, redox homeostasis, as well as intracellular metabolite transfer and signaling. We review here the current status of the different co-existing modes of biogenesis of peroxisomal membrane proteins demonstrating the fascinating adaptability in their targeting and sorting pathways. While earlier studies focused on peroxisomes as autonomous organelles, the necessity of the ER and potentially even mitochondria as sources of peroxisomal membrane proteins and lipids has come to light in recent years. Additionally, the intimate physical juxtaposition of peroxisomes with other organelles has transitioned from being viewed as random encounters to a growing appreciation of the expanding roles of such inter-organellar membrane contact sites in metabolic and regulatory functions. Peroxisomal quality control mechanisms have also come of age with a variety of mechanisms operating both during biogenesis and in the cellular response to environmental cues.
Collapse
Affiliation(s)
- Jean-Claude Farré
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, CA, USA
| | - Shanmuga S Mahalingam
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, CA, USA
| | - Marco Proietto
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, CA, USA
| | - Suresh Subramani
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, CA, USA
| |
Collapse
|
138
|
Interactions in the ESCRT-III network of the yeast Saccharomyces cerevisiae. Curr Genet 2018; 65:607-619. [PMID: 30506264 DOI: 10.1007/s00294-018-0915-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/28/2018] [Accepted: 11/28/2018] [Indexed: 01/27/2023]
Abstract
Here, we examine the genetic interactions between ESCRT-III mutations in the yeast Saccharomyces cerevisiae. From the obtained interaction network, we make predictions about alternative ESCRT-III complexes. By the successful generation of an octuple deletion strain using the CRISPR/Cas9 technique, we demonstrate for the first time that ESCRT-III activity as a whole is not essential for the life of a yeast cell. Endosomal sorting complex required for transport (ESCRT)-III proteins are membrane remodeling factors involved in a multitude of cellular processes. There are eight proteins in yeast with an ESCRT-III domain. It is not clear whether the diverse ESCRT-III functions are fulfilled by a single ESCRT-III complex or by different complexes with distinct composition. Genetic interaction studies may provide a hint on the existence of alternative complexes. We performed a genetic mini-array screen by analyzing the growth phenotypes of all pairwise combinations of ESCRT-III deletion mutations under different stress conditions. Our analysis is in line with previous data pointing to a complex containing Did2/CHMP1 and Ist1/IST1. In addition, we provide evidence for the existence of a novel complex consisting of Did2/CHMP1 and Vps2/CHMP2. Some of the interactions on Congo red plates could be explained by effects of ESCRT-III mutations on Rim101 signaling.
Collapse
|
139
|
Endosomal trafficking of yeast membrane proteins. Biochem Soc Trans 2018; 46:1551-1558. [PMID: 30381337 DOI: 10.1042/bst20180258] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/01/2018] [Accepted: 09/14/2018] [Indexed: 01/19/2023]
Abstract
Various membrane trafficking pathways transport molecules through the endosomal system of eukaryotic cells, where trafficking decisions control the localisation and activity of a diverse repertoire of membrane protein cargoes. The budding yeast Saccharomyces cerevisiae has been used to discover and define many mechanisms that regulate conserved features of endosomal trafficking. Internalised surface membrane proteins first localise to endosomes before sorting to other compartments. Ubiquitination of endosomal membrane proteins is a signal for their degradation. Ubiquitinated cargoes are recognised by the endosomal sorting complex required for transport (ESCRT) apparatus, which mediate sorting through the multivesicular body pathway to the lysosome for degradation. Proteins that are not destined for degradation can be recycled to other intracellular compartments, such as the Golgi and the plasma membrane. In this review, we discuss recent developments elucidating the mechanisms that drive membrane protein degradation and recycling pathways in yeast.
Collapse
|
140
|
McCullough J, Frost A, Sundquist WI. Structures, Functions, and Dynamics of ESCRT-III/Vps4 Membrane Remodeling and Fission Complexes. Annu Rev Cell Dev Biol 2018; 34:85-109. [PMID: 30095293 PMCID: PMC6241870 DOI: 10.1146/annurev-cellbio-100616-060600] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRT) pathway mediates cellular membrane remodeling and fission reactions. The pathway comprises five core complexes: ALIX, ESCRT-I, ESCRT-II, ESCRT-III, and Vps4. These soluble complexes are typically recruited to target membranes by site-specific adaptors that bind one or both of the early-acting ESCRT factors: ALIX and ESCRT-I/ESCRT-II. These factors, in turn, nucleate assembly of ESCRT-III subunits into membrane-bound filaments that recruit the AAA ATPase Vps4. Together, ESCRT-III filaments and Vps4 remodel and sever membranes. Here, we review recent advances in our understanding of the structures, activities, and mechanisms of the ESCRT-III and Vps4 machinery, including the first high-resolution structures of ESCRT-III filaments, the assembled Vps4 enzyme in complex with an ESCRT-III substrate, the discovery that ESCRT-III/Vps4 complexes can promote both inside-out and outside-in membrane fission reactions, and emerging mechanistic models for ESCRT-mediated membrane fission.
Collapse
Affiliation(s)
- John McCullough
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA;
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Wesley I Sundquist
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA;
| |
Collapse
|
141
|
Agudo-Canalejo J, Lipowsky R. Domes and cones: Adhesion-induced fission of membranes by ESCRT proteins. PLoS Comput Biol 2018; 14:e1006422. [PMID: 30130367 PMCID: PMC6118396 DOI: 10.1371/journal.pcbi.1006422] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/31/2018] [Accepted: 08/08/2018] [Indexed: 01/07/2023] Open
Abstract
ESCRT proteins participate in the fission step of exocytic membrane budding, by assisting in the closure and scission of the membrane neck that connects the nascent bud to the plasma membrane. However, the precise mechanism by which the proteins achieve this so-called reverse-topology membrane scission remains to be elucidated. One mechanism is described by the dome model, which postulates that ESCRT-III proteins assemble in the shape of a hemispherical dome at the location of the neck, and guide the closure of this neck via membrane–protein adhesion. A different mechanism is described by the flattening cone model, in which the ESCRT-III complex first assembles at the neck in the shape of a cone, which then flattens leading to neck closure. Here, we use the theoretical framework of curvature elasticity and membrane–protein adhesion to quantitatively describe and compare both mechanisms. This comparison shows that the minimal adhesive strength of the membrane–protein interactions required for scission is much lower for cones than for domes, and that the geometric constraints on the shape of the assembly required to induce scission are more stringent for domes than for cones. Finally, we compute for the first time the adhesion-induced constriction forces exerted by the ESCRT assemblies onto the membrane necks. These forces are higher for cones and of the order of 100 pN. Membrane fission is a crucial step in many biological processes ranging from cell division to viral budding. During fission, the membrane forms a narrow neck that is subsequently cleaved by proteins. ESCRT proteins initiate this process by forming supramolecular assemblies that adhere to the cytosolic face of the membranes. However, how these assemblies achieve the closure and scission of membrane necks is still a matter of controversy. Here, we elucidate and compare the two most prominent mechanisms that have been proposed for ESCRT-induced membrane fission. In both mechanisms, the ESCRT molecules form assemblies that adhere to the open neck of the membrane bud. In the first mechanism, these assemblies grow in a dome-like shape, whereas in the second mechanism the assemblies take a cone-like shape and flatten as they grow. Our computational study shows that the minimal strength of membrane–protein adhesion necessary for cone-induced fission is lower than for dome-induced fission, and that cones generate higher constriction forces of the order of 100 pN onto the membrane necks.
Collapse
Affiliation(s)
- Jaime Agudo-Canalejo
- Theory & Bio-Systems Department, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Oxford, United Kingdom
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (JAC); (RL)
| | - Reinhard Lipowsky
- Theory & Bio-Systems Department, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- * E-mail: (JAC); (RL)
| |
Collapse
|
142
|
Goliand I, Adar-Levor S, Segal I, Nachmias D, Dadosh T, Kozlov MM, Elia N. Resolving ESCRT-III Spirals at the Intercellular Bridge of Dividing Cells Using 3D STORM. Cell Rep 2018; 24:1756-1764. [PMID: 30110633 DOI: 10.1016/j.celrep.2018.07.051] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 04/10/2018] [Accepted: 07/16/2018] [Indexed: 11/26/2022] Open
Abstract
The ESCRT machinery mediates membrane fission in a variety of processes in cells. According to current models, ESCRT-III proteins drive membrane fission by assembling into helical filaments on membranes. Here, we used 3D STORM imaging of endogenous ESCRT-III component IST1 to reveal the evolution of the structural organization of ESCRT-III in mammalian cytokinetic abscission. Using this approach, ESCRT-III ring and spiral assemblies were resolved and characterized at different stages of abscission. Visualization of IST1 structures in cells lacking the microtubule-severing enzyme spastin and in cells depleted of specific ESCRT-III components or the ATPase VPS4 demonstrated the contribution of these components to the organization and function of ESCRTs in cells. This work provides direct evidence that ESCRT-III proteins form helical filaments to mediate their function in cells and raises new mechanistic scenarios for ESCRT-driven cytokinetic abscission.
Collapse
Affiliation(s)
- Inna Goliand
- Department of Life Sciences and NIBN, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Shai Adar-Levor
- Department of Life Sciences and NIBN, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Inbar Segal
- Department of Life Sciences and NIBN, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Dikla Nachmias
- Department of Life Sciences and NIBN, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Faculty of Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Natalie Elia
- Department of Life Sciences and NIBN, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
143
|
De Franceschi N, Alqabandi M, Miguet N, Caillat C, Mangenot S, Weissenhorn W, Bassereau P. The ESCRT protein CHMP2B acts as a diffusion barrier on reconstituted membrane necks. J Cell Sci 2018; 132:jcs.217968. [PMID: 29967034 DOI: 10.1242/jcs.217968] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/25/2018] [Indexed: 01/23/2023] Open
Abstract
Endosomal sorting complexes required for transport (ESCRT)-III family proteins catalyze membrane remodeling processes that stabilize and constrict membrane structures. It has been proposed that stable ESCRT-III complexes containing CHMP2B could establish diffusion barriers at the post-synaptic spine neck. In order to better understand this process, we developed a novel method based on fusion of giant unilamellar vesicles to reconstitute ESCRT-III proteins inside GUVs, from which membrane nanotubes are pulled. The new assay ensures that ESCRT-III proteins polymerize only when they become exposed to physiologically relevant membrane topology mimicking the complex geometry of post-synaptic spines. We establish that CHMP2B, both full-length and with a C-terminal deletion (ΔC), preferentially binds to membranes containing phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Moreover, we show that CHMP2B preferentially accumulates at the neck of membrane nanotubes, and provide evidence that CHMP2B-ΔC prevents the diffusion of PI(4,5)P2 lipids and membrane-bound proteins across the tube neck. This indicates that CHMP2B polymers formed at a membrane neck may function as a diffusion barrier, highlighting a potential important function of CHMP2B in maintaining synaptic spine structures.
Collapse
Affiliation(s)
- Nicola De Franceschi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France.,Sorbonne Université, 75005, Paris, France
| | - Maryam Alqabandi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France.,Sorbonne Université, 75005, Paris, France
| | - Nolwenn Miguet
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71, avenue des Martyrs, 38000 Grenoble, France
| | - Christophe Caillat
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71, avenue des Martyrs, 38000 Grenoble, France
| | - Stephanie Mangenot
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France.,Sorbonne Université, 75005, Paris, France
| | - Winfried Weissenhorn
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CEA, CNRS, 71, avenue des Martyrs, 38000 Grenoble, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France .,Sorbonne Université, 75005, Paris, France
| |
Collapse
|
144
|
Johnson DS, Bleck M, Simon SM. Timing of ESCRT-III protein recruitment and membrane scission during HIV-1 assembly. eLife 2018; 7:36221. [PMID: 29972351 PMCID: PMC6080951 DOI: 10.7554/elife.36221] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/02/2018] [Indexed: 12/20/2022] Open
Abstract
The Endosomal Sorting Complexes Required for Transport III (ESCRT-III) proteins are critical for cellular membrane scission processes with topologies inverted relative to clathrin-mediated endocytosis. Some viruses appropriate ESCRT-IIIs for their release. By imaging single assembling viral-like particles of HIV-1, we observed that ESCRT-IIIs and the ATPase VPS4 arrive after most of the virion membrane is bent, linger for tens of seconds, and depart ~20 s before scission. These observations suggest that ESCRT-IIIs are recruited by a combination of membrane curvature and the late domains of the HIV-1 Gag protein. ESCRT-IIIs may pull the neck into a narrower form but must leave to allow scission. If scission does not occur within minutes of ESCRT departure, ESCRT-IIIs and VPS4 are recruited again. This mechanistic insight is likely relevant for other ESCRT-dependent scission processes including cell division, endosome tubulation, multivesicular body and nuclear envelope formation, and secretion of exosomes and ectosomes.
Collapse
Affiliation(s)
- Daniel S Johnson
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, United States.,Department of Physics and Astronomy, Hofstra University, Hempstead, United States
| | - Marina Bleck
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, United States
| | - Sanford M Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, United States
| |
Collapse
|
145
|
Crespo-Yàñez X, Aguilar-Gurrieri C, Jacomin AC, Journet A, Mortier M, Taillebourg E, Soleilhac E, Weissenhorn W, Fauvarque MO. CHMP1B is a target of USP8/UBPY regulated by ubiquitin during endocytosis. PLoS Genet 2018; 14:e1007456. [PMID: 29933386 PMCID: PMC6033466 DOI: 10.1371/journal.pgen.1007456] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 07/05/2018] [Accepted: 05/30/2018] [Indexed: 11/29/2022] Open
Abstract
Integration and down-regulation of cell growth and differentiation signals rely on plasma membrane receptor endocytosis and sorting towards either recycling vesicles or degradative lysosomes via multivesicular bodies (MVB). In this process, the endosomal sorting complex-III required for transport (ESCRT-III) controls membrane deformation and scission triggering intraluminal vesicle (ILV) formation at early endosomes. Here, we show that the ESCRT-III member CHMP1B can be ubiquitinated within a flexible loop known to undergo conformational changes during polymerization. We demonstrate further that CHMP1B is deubiquitinated by the ubiquitin specific protease USP8 (syn. UBPY) and found fully devoid of ubiquitin in a ~500 kDa large complex that also contains its ESCRT-III partner IST1. Moreover, EGF stimulation induces the rapid and transient accumulation of ubiquitinated forms of CHMP1B on cell membranes. Accordingly, CHMP1B ubiquitination is necessary for CHMP1B function in both EGF receptor trafficking in human cells and wing development in Drosophila. Based on these observations, we propose that CHMP1B is dynamically regulated by ubiquitination in response to EGF and that USP8 triggers CHMP1B deubiquitination possibly favoring its subsequent assembly into a membrane-associated ESCRT-III polymer. In multicellular organisms, the interpretation and transmission of cell growth and differentiation signals strongly rely on plasma membrane receptors. Once activated by their ligands, these receptors activate downstream signaling cascades and are rapidly internalized into intracellular vesicles that fuse inside the cell to form the endosomal compartment. From there, the receptors are sorted towards either recycling vesicles or degradative lysosomes via multivesicular bodies. Receptors sorting therefore plays a crucial role in the integration and regulation of intracellular signals during development and numerous physio-pathological processes. It requires extensive membrane remodeling and scission events at the level of the endosomal compartment by so-called ESCRT proteins, including CHMP1B. In this study, we provide evidence for dynamic regulation of CHMP1B function and subcellular localization by ubiquitin linkage. We also show the contribution of the ubiquitin specific protease USP8 in this regulation, which is a known actor of intracellular trafficking and Cushing’s disease.
Collapse
Affiliation(s)
- Xènia Crespo-Yàñez
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
| | - Carmen Aguilar-Gurrieri
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France
| | - Anne-Claire Jacomin
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
| | - Agnès Journet
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
| | - Magda Mortier
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
| | - Emmanuel Taillebourg
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
| | - Emmanuelle Soleilhac
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
| | - Winfried Weissenhorn
- Institut de Biologie Structurale (IBS), Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France
| | - Marie-Odile Fauvarque
- Institut de Biosciences et Biotechnologies de Grenoble (BIG), Univ. Grenoble Alpes, INSERM U1038, CEA, Grenoble, France
- * E-mail:
| |
Collapse
|
146
|
Mast FD, Herricks T, Strehler KM, Miller LR, Saleem RA, Rachubinski RA, Aitchison JD. ESCRT-III is required for scissioning new peroxisomes from the endoplasmic reticulum. J Cell Biol 2018; 217:2087-2102. [PMID: 29588378 PMCID: PMC5987711 DOI: 10.1083/jcb.201706044] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/23/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022] Open
Abstract
Dynamic control of peroxisome proliferation is integral to the peroxisome's many functions. The endoplasmic reticulum (ER) serves as a source of preperoxisomal vesicles (PPVs) that mature into peroxisomes during de novo peroxisome biogenesis and support growth and division of existing peroxisomes. However, the mechanism of PPV formation and release from the ER remains poorly understood. In this study, we show that endosomal sorting complexes required for transport (ESCRT)-III are required to release PPVs budding from the ER into the cytosol. Absence of ESCRT-III proteins impedes de novo peroxisome formation and results in an aberrant peroxisome population in vivo. Using a cell-free PPV budding assay, we show that ESCRT-III proteins Vps20 and Snf7 are necessary to release PPVs from the ER. ESCRT-III is therefore a positive effector of membrane scission for vesicles budding both away from and toward the cytosol. These findings have important implications for the evolutionary timing of emergence of peroxisomes and the rest of the internal membrane architecture of the eukaryotic cell.
Collapse
Affiliation(s)
- Fred D. Mast
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Thurston Herricks
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Kathleen M. Strehler
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Leslie R. Miller
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Ramsey A. Saleem
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | | | - John D. Aitchison
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
147
|
Egea-Jimenez AL, Zimmermann P. Phospholipase D and phosphatidic acid in the biogenesis and cargo loading of extracellular vesicles. J Lipid Res 2018; 59:1554-1560. [PMID: 29853529 DOI: 10.1194/jlr.r083964] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/09/2018] [Indexed: 12/30/2022] Open
Abstract
Extracellular vesicles released by viable cells (exosomes and microvesicles) have emerged as important organelles supporting cell-cell communication. Because of their potential therapeutic significance, important efforts are being made toward characterizing the contents of these vesicles and the mechanisms that govern their biogenesis. It has been recently demonstrated that the lipid modifying enzyme, phospholipase D (PLD)2, is involved in exosome production and acts downstream of the small GTPase, ARF6. This review aims to recapitulate our current knowledge of the role of PLD2 and its product, phosphatidic acid, in the biogenesis of exosomes and to propose hypotheses for further investigation of a possible central role of these molecules in the biology of these organelles.
Collapse
Affiliation(s)
- Antonio Luis Egea-Jimenez
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe labellisée LIGUE 2018, Aix-Marseille Université, Marseille F-13284, France and Inserm U1068, Institut Paoli-Calmettes, and CNRS UMR7258, Marseille F-13009, France
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe labellisée LIGUE 2018, Aix-Marseille Université, Marseille F-13284, France and Inserm U1068, Institut Paoli-Calmettes, and CNRS UMR7258, Marseille F-13009, France; Department of Human Genetics, University of Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
148
|
Snead WT, Stachowiak JC. Structure Versus Stochasticity-The Role of Molecular Crowding and Intrinsic Disorder in Membrane Fission. J Mol Biol 2018; 430:2293-2308. [PMID: 29627460 DOI: 10.1016/j.jmb.2018.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 01/07/2023]
Abstract
Cellular membranes must undergo remodeling to facilitate critical functions including membrane trafficking, organelle biogenesis, and cell division. An essential step in membrane remodeling is membrane fission, in which an initially continuous membrane surface is divided into multiple, separate compartments. The established view has been that membrane fission requires proteins with conserved structural features such as helical scaffolds, hydrophobic insertions, and polymerized assemblies. In this review, we discuss these structure-based fission mechanisms and highlight recent findings from several groups that support an alternative, structure-independent mechanism of membrane fission. This mechanism relies on lateral collisions among crowded, membrane-bound proteins to generate sufficient steric pressure to drive membrane vesiculation. As a stochastic process, this mechanism contrasts with the paradigm that deterministic protein structures are required to drive fission, raising the prospect that many more proteins may participate in fission than previously thought. Paradoxically, our recent work suggests that intrinsically disordered domains may be among the most potent drivers of membrane fission, owing to their large hydrodynamic radii and substantial chain entropy. This stochastic view of fission also suggests new roles for the structure-based fission proteins. Specifically, we hypothesize that in addition to driving fission directly, the canonical fission machines may facilitate the enrichment and organization of bulky disordered protein domains in order to promote membrane fission by locally amplifying protein crowding.
Collapse
Affiliation(s)
- Wilton T Snead
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jeanne C Stachowiak
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
149
|
Increasing Diversity of Biological Membrane Fission Mechanisms. Trends Cell Biol 2018; 28:274-286. [DOI: 10.1016/j.tcb.2017.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/06/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022]
|
150
|
McMillan BJ, Tibbe C, Drabek AA, Seegar TCM, Blacklow SC, Klein T. Structural Basis for Regulation of ESCRT-III Complexes by Lgd. Cell Rep 2018; 19:1750-1757. [PMID: 28564595 DOI: 10.1016/j.celrep.2017.05.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/04/2017] [Accepted: 05/07/2017] [Indexed: 11/17/2022] Open
Abstract
The ESCRT-III complex induces outward membrane budding and fission through homotypic polymerization of its core component Shrub/CHMP4B. Shrub activity is regulated by its direct interaction with a protein called Lgd in flies, or CC2D1A or B in humans. Here, we report the structural basis for this interaction and propose a mechanism for regulation of polymer assembly. The isolated third DM14 repeat of Lgd binds Shrub, and an Lgd fragment containing only this DM14 repeat and its C-terminal C2 domain is sufficient for in vivo function. The DM14 domain forms a helical hairpin with a conserved, positively charged tip, that, in the structure of a DM14 domain-Shrub complex, occupies a negatively charged surface of Shrub that is otherwise used for homopolymerization. Lgd mutations at this interface disrupt its function in flies, confirming functional importance. Together, these data argue that Lgd regulates ESCRT activity by controlling access to the Shrub self-assembly surface.
Collapse
Affiliation(s)
- Brian J McMillan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Christine Tibbe
- Institute of Genetics, Heinrich-Heine-University, Dusseldorf 40225, Germany
| | - Andrew A Drabek
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Tom C M Seegar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Dana Farber Cancer Institute, Department of Cancer Biology, Boston, MA 02215, USA.
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-University, Dusseldorf 40225, Germany.
| |
Collapse
|