1
|
Liao Z, Zeng J, Lin A, Zou Y, Zhou Z. Pre-treated mesenchymal stem cell-derived exosomes: A new perspective for accelerating spinal cord injury repair. Eur J Pharmacol 2025; 992:177349. [PMID: 39921061 DOI: 10.1016/j.ejphar.2025.177349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Spinal cord injury (SCI) is a devastating event for the central nervous system (CNS), often resulting in the loss of sensory and motor functions. It profoundly affects both the physiological and psychological well-being of patients, reducing their quality of life while also imposing significant economic pressure on families and the healthcare system. Due to the complex pathophysiology of SCI, effective treatments for promoting recovery remain scarce. Mesenchymal stem cell-derived exosomes (MSC-Exos) offer advantages such as low immunogenicity, good biocompatibility, and the ability to cross the blood-spinal cord barrier (BSCB). In preclinical studies, they have progressively shown efficacy in promoting SCI repair and functional recovery. However, the low yield and insufficient targeting of MSC-Exos limit their therapeutic efficacy. Currently, genetic engineering and other preprocessing techniques are being employed to optimize both the yield and functional properties of exosomes, thereby enhancing their therapeutic potential. Therefore, this paper provides an overview of the pathophysiology of SCI and the biogenesis of exosomes. It also summarizes current approaches to optimizing exosome performance. Additionally, it details the mechanisms through which optimized exosomes provide neuroprotection and explores the potential of combined treatments involving MSC-Exos and hydrogels.
Collapse
Affiliation(s)
- Zhiqiang Liao
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Junjian Zeng
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Aiqing Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Yu Zou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Zhidong Zhou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
2
|
Naskar S, Merino A, Espadas J, Singh J, Roux A, Colom A, Low HH. Mechanism for Vipp1 spiral formation, ring biogenesis, and membrane repair. Nat Struct Mol Biol 2025; 32:571-584. [PMID: 39528797 PMCID: PMC11919738 DOI: 10.1038/s41594-024-01401-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 09/11/2024] [Indexed: 11/16/2024]
Abstract
The ESCRT-III-like protein Vipp1 couples filament polymerization with membrane remodeling. It assembles planar sheets as well as 3D rings and helical polymers, all implicated in mitigating plastid-associated membrane stress. The architecture of Vipp1 planar sheets and helical polymers remains unknown, as do the geometric changes required to transition between polymeric forms. Here we show how cyanobacterial Vipp1 assembles into morphologically-related sheets and spirals on membranes in vitro. The spirals converge to form a central ring similar to those described in membrane budding. Cryo-EM structures of helical filaments reveal a close geometric relationship between Vipp1 helical and planar lattices. Moreover, the helical structures reveal how filaments twist-a process required for Vipp1, and likely other ESCRT-III filaments, to transition between planar and 3D architectures. Overall, our results provide a molecular model for Vipp1 ring biogenesis and a mechanism for Vipp1 membrane stabilization and repair, with implications for other ESCRT-III systems.
Collapse
Affiliation(s)
- Souvik Naskar
- Department of Infectious Disease, Imperial College, London, UK
| | - Andrea Merino
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country, Leioa, Spain
| | - Javier Espadas
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| | - Jayanti Singh
- Department of Infectious Disease, Imperial College, London, UK
| | - Aurelien Roux
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| | - Adai Colom
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country, Leioa, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | - Harry H Low
- Department of Infectious Disease, Imperial College, London, UK.
| |
Collapse
|
3
|
Pan S, Gries K, Engel BD, Schroda M, Haselwandter CA, Scheuring S. The cyanobacterial protein VIPP1 forms ESCRT-III-like structures on lipid bilayers. Nat Struct Mol Biol 2025; 32:543-554. [PMID: 39060677 PMCID: PMC11762370 DOI: 10.1038/s41594-024-01367-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 07/03/2024] [Indexed: 07/28/2024]
Abstract
The biogenesis and maintenance of thylakoid membranes require vesicle-inducing protein in plastids 1 (VIPP1). VIPP1 is a member of the endosomal sorting complex required for transport-III (ESCRT-III) superfamily, whose members form diverse filament-based supramolecular structures that facilitate membrane deformation and fission. VIPP1 cryo-electron microscopy (EM) structures in solution revealed helical rods and baskets of stacked rings, with amphipathic membrane-binding domains in the lumen. However, how VIPP1 interacts with membranes remains largely unknown. Here, using high-speed atomic force microscopy (HS-AFM), we show that VIPP1 assembles into right-handed chiral spirals and regular polygons on supported lipid bilayers via ESCRT-III-like filament assembly and dynamics. VIPP1 filaments grow clockwise into spirals through polymerization at a ring-shaped central polymerization hub, and into polygons through clockwise polymerization at the sector peripheries. Interestingly, VIPP1 initially forms Archimedean spirals, which upon maturation transform into logarithmic spirals through lateral annealing of strands to the outermore low-curvature spiral turns.
Collapse
Affiliation(s)
- Sichen Pan
- Weill Cornell Medicine, Department of Anesthesiology, New York, NY, USA
| | - Karin Gries
- Molecular Biotechnology and Systems Biology, RPTU Kaiserslautern-Landau, Kaiserslautern, Germany
| | | | - Michael Schroda
- Molecular Biotechnology and Systems Biology, RPTU Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Christoph A Haselwandter
- Department of Physics and Astronomy, University of Southern California, Los Angeles, CA, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Simon Scheuring
- Weill Cornell Medicine, Department of Anesthesiology, New York, NY, USA.
- Weill Cornell Medicine, Department of Physiology and Biophysics, New York, NY, USA.
| |
Collapse
|
4
|
Henn D, Yang X, Li M. Lysosomal quality control Review. Autophagy 2025:1-20. [PMID: 39968899 DOI: 10.1080/15548627.2025.2469206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025] Open
Abstract
Healthy cells need functional lysosomes to degrade cargo delivered by autophagy and endocytosis. Defective lysosomes can lead to severe conditions such as lysosomal storage diseases (LSDs) and neurodegeneration. To maintain lysosome integrity and functionality, cells have evolved multiple quality control pathways corresponding to different types of stress and damage. These can be divided into five levels: regulation, reformation, repair, removal, and replacement. The different levels of lysosome quality control often work together to maintain the integrity of the lysosomal network. This review summarizes the different quality control pathways and discusses the less-studied area of lysosome membrane protein regulation and degradation, highlighting key unanswered questions in the field.Abbreviation: ALR: autophagic lysosome reformation; CASM: conjugation of ATG8 to single membranes: ER: endoplasmic reticulum; ESCRT: endosomal sorting complexes required for transport; ILF: intralumenal fragment; LSD: lysosomal storage disease; LYTL: lysosomal tubulation/sorting driven by LRRK2; PITT: phosphoinositide-initiated membrane tethering and lipid transport; PE: phosphatidylethanolamine; PLR: phagocytic lysosome reformation; PS: phosphatidylserine; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns4P: phosphatidylinositol-4-phosphate; PtdIns(4,5)P2: phosphatidylinositol-4,5-bisphosphate; V-ATPase: vacuolar-type H+-translocating ATPase.
Collapse
Affiliation(s)
- Danielle Henn
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Xi Yang
- Department of Biological Sciences, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Ming Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Nachmias D, Frohn BP, Sachse C, Mizrahi I, Elia N. ESCRTs - a multi-purpose membrane remodeling device encoded in all life forms. Trends Microbiol 2025:S0966-842X(25)00008-3. [PMID: 39979199 DOI: 10.1016/j.tim.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/22/2025]
Abstract
The ESCRT (endosomal sorting complexes required for transport) membrane remodeling complex, found across all life forms, exhibits a versatility that transcends evolutionary boundaries. From orchestrating the constriction of micron-wide tubes in cell division to facilitating the budding of 50 nm vesicles in receptor degradation, ESCRTs perform diverse functions in animal cells. However, the basis of this functional diversity remains enigmatic. While extensively studied in eukaryotes, the role of ESCRTs in prokaryotes is only beginning to emerge. This review synthesizes data on ESCRT systems across the tree of life, focusing on microorganisms and drawing parallels to their functions in human cells. This comparative approach highlights the remarkable plasticity of the ESCRT system across functional, structural, and genomic levels in both prokaryotes and eukaryotes. This integrated knowledge supports a model in which the ESCRT system evolved as a multipurpose membrane remodeling tool, adaptable to specific functions within and across organisms. Our review not only underscores the significance of ESCRTs in microorganisms but also paves the way for exciting avenues of research into the intricacies of cellular membrane dynamics, offering valuable insights into the evolution of cellular complexity across diverse organisms and ecosystems.
Collapse
Affiliation(s)
- Dikla Nachmias
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Béla P Frohn
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Carsten Sachse
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany; Department of Biology, Heinrich Heine University, Dusseldorf, Germany
| | - Itzhak Mizrahi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; The Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Natalie Elia
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
6
|
Souza DP, Espadas J, Chaaban S, Moody ERR, Hatano T, Balasubramanian M, Williams TA, Roux A, Baum B. Asgard archaea reveal the conserved principles of ESCRT-III membrane remodeling. SCIENCE ADVANCES 2025; 11:eads5255. [PMID: 39919172 PMCID: PMC11804906 DOI: 10.1126/sciadv.ads5255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025]
Abstract
ESCRT-III proteins assemble into composite polymers that undergo stepwise changes in composition and structure to deform membranes across the tree of life. Here, using a phylogenetic analysis, we demonstrate that the two endosomal sorting complex required for transport III (ESCRT-III) proteins present in eukaryote's closest Asgard archaeal relatives are evolutionarily related to the B- and A-type eukaryotic paralogs that initiate and execute membrane remodeling, respectively. We show that Asgard ESCRT-IIIB assembles into parallel arrays on planar membranes to initiate membrane deformation, from where it recruits ESCRT-IIIA to generate composite polymers. Last, we show that Asgard ESCRT-IIIA is able to remodel membranes into tubes as a likely prelude to scission. Together, these data reveal a set of conserved principles governing ESCRT-III-dependent membrane remodeling that first emerged in a two-component ESCRT-III system in archaea.
Collapse
Affiliation(s)
| | - Javier Espadas
- Department of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Sami Chaaban
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Edmund R. R. Moody
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
| | - Tomoyuki Hatano
- Centre for Mechanochemical Cell Biology, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Mohan Balasubramanian
- Centre for Mechanochemical Cell Biology, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Tom A. Williams
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Buzz Baum
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| |
Collapse
|
7
|
Melnikov N, Junglas B, Halbi G, Nachmias D, Zerbib E, Gueta N, Upcher A, Zalk R, Sachse C, Bernheim-Groswasser A, Elia N. The Asgard archaeal ESCRT-III system forms helical filaments and remodels eukaryotic-like membranes. EMBO J 2025; 44:665-681. [PMID: 39753954 PMCID: PMC11791191 DOI: 10.1038/s44318-024-00346-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 02/05/2025] Open
Abstract
The ESCRT machinery mediates membrane remodeling in numerous processes in cells including cell division and nuclear membrane reformation. The identification of ESCRT homologs in Asgard archaea, currently considered the closest prokaryotic relative of eukaryotes, implies a role for ESCRTs in the membrane remodeling processes that occurred during eukaryogenesis. Yet, the function of these distant ESCRT homologs is mostly unresolved. Here we show that Asgard ESCRT-III proteins of the Lokiarcheota self-assemble into helical filaments, a hallmark of the ESCRT system. We determined the cryo-EM structure of the filaments at 3.6 Å resolution and found that they share features of bacterial and eukaryotic ESCRT-III assemblies. Markedly, Asgard ESCRT-III filaments bound and deformed eukaryotic-like membrane vesicles. Oligonucleotides facilitated the assembly of ESCRT-III filaments and tuned the extent of membrane remodeling. The ability of Asgard archaeal ESCRTs to remodel eukaryotic-like membranes, which are fundamentally different from archaeal membranes, and the structural properties of these proteins places them at the junction between prokaryotes and eukaryotes.
Collapse
Affiliation(s)
- Nataly Melnikov
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Benedikt Junglas
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Gal Halbi
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Dikla Nachmias
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Erez Zerbib
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Noam Gueta
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Alexander Upcher
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Carsten Sachse
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, 52425, Jülich, Germany.
- Department of Biology, Heinrich Heine University, Universitätsstr. 1, 40225, Düsseldorf, Germany.
| | - Anne Bernheim-Groswasser
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Natalie Elia
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
| |
Collapse
|
8
|
Huang X, Zhang J, Xu C, Cao R, Jiang P, Ji X, Wang W, Huang Z, Han P. Vps4a Mediates a Unified Membrane Repair Machinery to Attenuate Ischemia/Reperfusion Injury. Circ Res 2025; 136:279-296. [PMID: 39764631 DOI: 10.1161/circresaha.124.325290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Cardiac ischemia/reperfusion disrupts plasma membrane integrity and induces various types of programmed cell death. The ESCRT (endosomal sorting complex required for transport) proteins, particularly AAA-ATPase Vps4a (vacuolar protein sorting 4a), play an essential role in the surveillance of membrane integrity. However, the role of ESCRT proteins in the context of cardiac injury remains unclear. METHODS We simultaneously visualized the formation of membrane blebs and the subcellular translocation of Vps4a during a variety of cell death programs in primary cardiomyocytes. Vps4a cardiomyocyte-specific knockout and overexpression mice were generated and characterized. In vivo and ex vivo surgeries were performed to determine the effects of altered Vps4a expression levels on plasma membrane repair and cell survival. Given the role of Ripk3 (receptor-interacting kinase 3)-mediated pore formation in regulating cell membrane integrity, hearts from Ripk3 and Vps4a double-knockout mice were examined. The sequential recruitment of upstream ESCRT components that promote the translocation of Vps4a to injured sites was also assessed using genetic gain- and loss-of-function approaches. Finally, we overexpressed a mutated form of Vps4a with defective ATPase activity and investigated its function during cardiomyocyte membrane repair. RESULTS Ischemia/reperfusion stimulation or forced induction of apoptosis, necroptosis, and pyroptosis in primary cardiomyocytes leads to membrane blebbing and the exposure of phosphatidylserine to the extracellular space. In response to injury, Vps4a promptly translocates to injured sites to reseal damaged membranes. Vps4a gain- and loss-of-function in the postnatal stage minimally affects cardiac structure formation and function. However, in the context of ischemia/reperfusion stimulation, overexpression of Vps4a protects cardiomyocytes against injury, whereas Vps4a-deficient hearts are more susceptible to cell damage. Additionally, Ripk3 deletion abrogates the detrimental effects of Vps4a deficiency during ischemia/reperfusion injury, and the Ca2+-Alix-Ist1 axis plays an essential role in recruiting Vps4a to the injured site. Mechanistically, Vps4a promotes the shedding of plasma membrane blebs to restrict permeability to the extracellular environment, and the surveillance of membrane integrity requires the ATPase activity of Vps4a. CONCLUSIONS These results demonstrate that Vps4a-mediated plasma membrane repair is an intrinsic cell protection machinery that antagonizes cardiac ischemia/reperfusion injury, and our findings may contribute to the development of therapeutic strategies towards attenuating cardiac injury.
Collapse
Affiliation(s)
- Xiaozhi Huang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Jiayin Zhang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Chen Xu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Ranran Cao
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Peijun Jiang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Xue Ji
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Wenyi Wang
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Zhishan Huang
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| | - Peidong Han
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, China (X.H., J.Z., C.X., R.C., P.J., X.J., P.H.)
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China (X.H., J.Z., C.X., R.C., P.J., X.J., W.W., Z.H., P.H.)
| |
Collapse
|
9
|
Ovčar A, Kovačič B. Biogenesis of Extracellular Vesicles (EVs) and the Potential Use of Embryo-Derived EVs in Medically Assisted Reproduction. Int J Mol Sci 2024; 26:42. [PMID: 39795901 PMCID: PMC11719982 DOI: 10.3390/ijms26010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-bound particles released from cells that cannot replicate on their own, play a crucial role in intercellular communication, and are implicated in various physiological and pathological processes. Within the domain of embryo culture media research, extensive studies have been conducted to evaluate embryo viability by analyzing spent culture medium. Advanced methodologies such as metabolomic profiling, proteomic and genomic analyses, transcriptomic profiling, non-coding RNA assessments, and oxidative status measurements have been employed to further understand the molecular characteristics of embryos and improve selection criteria for successful implantation. In the field of EVs, only a limited number of studies have been conducted on embryo-conditioned medium, indicating a significant gap in knowledge regarding the potential role of EVs in embryo development and implantation. Therefore, this review aims to evaluate current research findings on EVs enriched from animal and human embryo spent medium. By unraveling the potential link between embryo-derived EVs and embryo selection in clinical settings, such research might enhance embryo-selection methods in assisted reproductive technologies, eventually increasing the success rates of fertility treatments and advancing our understanding of mechanisms underlying successful embryo development and implantation in humans.
Collapse
Affiliation(s)
| | - Borut Kovačič
- Department of Reproductive Medicine and Gynecological Endocrinology, University Medical Centre Maribor, 2000 Maribor, Slovenia;
| |
Collapse
|
10
|
De Lira Silva NS, Schenkman S. Biogenesis of EVs in Trypanosomatids. CURRENT TOPICS IN MEMBRANES 2024; 94:49-83. [PMID: 39370213 DOI: 10.1016/bs.ctm.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Trypanosomes are protozoan parasites responsible for human diseases such as Chagas disease, African trypanosomiasis, and leishmaniasis. These organisms' growth in various environments and exhibit multiple morphological stages, while adapting their surface components. They acquire and release materials extensively to get nutrients and manage interactions with the extracellular environment. They acquire and utilize proteins, lipids, and carbohydrates for growth via using membrane transport and endocytosis. Endocytosis takes place through distinct membrane areas known as the flagellar pocket and cytostome, depending on the parasite species and its developmental stage. Some forms establish a complex endocytic system to either store or break down the absorbed materials. In contrast, membrane transport facilitates the uptake of small molecules like amino acids, carbohydrates, and iron via particular receptors on the plasma membrane. Concurrently, these parasites secrete various molecules such as proteins, enzymes, nucleic acids, and glycoconjugates either in soluble form or enclosed in extracellular vesicles, which significantly contribute to their parasitic behavior. These activities require exocytosis through a secretory pathway in certain membrane domains such as the flagellum, flagellar pocket, and plasma membrane, which are controlled at various developmental stages. The main features of the endocytic and exocytic mechanisms, as well as the organelles involved, are discussed in this chapter along with their connection to the formation of exosomes and extracellular vesicles in the Tritryp species.
Collapse
Affiliation(s)
- Nadjania Saraiva De Lira Silva
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Sergio Schenkman
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Antimicrobial Resistance Institute of São Paulo (Aries), São Paulo, Brazil.
| |
Collapse
|
11
|
Liu YG, Jiang ST, Zhang JW, Zheng H, Zhang L, Zhao HT, Sang XT, Xu YY, Lu X. Role of extracellular vesicle-associated proteins in the progression, diagnosis, and treatment of hepatocellular carcinoma. Cell Biosci 2024; 14:113. [PMID: 39227992 PMCID: PMC11373138 DOI: 10.1186/s13578-024-01294-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, characterized by difficulties in early diagnosis, prone to distant metastasis, and high recurrence rates following surgery. Extracellular vesicles (EVs) are a class of cell-derived particles, including exosomes, characterized by a phospholipid bilayer. They serve as effective carriers for intercellular communication cargo, including proteins and nucleic acids, and are widely involved in tumor progression. They are being explored as potential tumor biomarkers and novel therapeutic avenues. We provide a brief overview of the biogenesis and characteristics of EVs to better understand their classification standards. The focus of this review is on the research progress of EV-associated proteins in the field of HCC. EV-associated proteins are involved in tumor growth and regulation in HCC, participate in intercellular communication within the tumor microenvironment (TME), and are implicated in events including angiogenesis and epithelial-mesenchymal transition (EMT) during tumor metastasis. In addition, EV-associated proteins show promising diagnostic efficacy for HCC. For the treatment of HCC, they also demonstrate significant potential including enhancing the efficacy of tumor vaccines, and as targeting cargo anchors. Facing current challenges, we propose the future directions of research in this field. Above all, research on EV-associated proteins offers the potential to enhance our comprehension of HCC and offer novel insights for developing new treatment strategies.
Collapse
Affiliation(s)
- Yao-Ge Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Shi-Tao Jiang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jun-Wei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Han Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Lei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Hai-Tao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xin-Ting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yi-Yao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China.
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China.
| |
Collapse
|
12
|
Park J, Kim J, Park H, Kim T, Lee S. ESCRT-III: a versatile membrane remodeling machinery and its implications in cellular processes and diseases. Anim Cells Syst (Seoul) 2024; 28:367-380. [PMID: 39070887 PMCID: PMC11275535 DOI: 10.1080/19768354.2024.2380294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
The endosomal sorting complexes required for transport (ESCRT) machinery is an evolutionarily conserved cytosolic protein complex that plays a crucial role in membrane remodeling and scission events across eukaryotes. Initially discovered for its function in multivesicular body (MVB) formation, the ESCRT complex has since been implicated in a wide range of membrane-associated processes, including endocytosis, exocytosis, cytokinesis, and autophagy. Recent advances have elucidated the ESCRT assembly pathway and highlighted the distinct functions of the various ESCRT complexes and their associated partners. Among the ESCRT complexes, ESCRT-III stands out as a critical player in membrane remodeling, with its subunits assembled into higher-order multimers capable of bending and severing membranes. This review focuses on the ESCRT-III complex, exploring its diverse functions in cellular processes beyond MVB biogenesis. We delve into the molecular mechanisms underlying ESCRT-III-mediated membrane remodeling and highlight its emerging roles in processes such as viral budding, autophagosome closure, and cytokinetic abscission. We also discuss the implications of ESCRT-III dysregulation in neurodegenerative diseases. The versatile membrane remodeling capabilities of ESCRT-III across diverse cellular processes underscore its importance in maintaining proper cellular function. Furthermore, we highlight the promising potential of ESCRT-III as a therapeutic target for neurodegenerative diseases, offering insights into the treatments of the diseases and the technical applications in related research fields.
Collapse
Affiliation(s)
- Jisoo Park
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Jongyoon Kim
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Hyungsun Park
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Taewan Kim
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Seongju Lee
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
- Department of Anatomy, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
13
|
Zhao W, Li K, Li L, Wang R, Lei Y, Yang H, Sun L. Mesenchymal Stem Cell-Derived Exosomes as Drug Delivery Vehicles in Disease Therapy. Int J Mol Sci 2024; 25:7715. [PMID: 39062956 PMCID: PMC11277139 DOI: 10.3390/ijms25147715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Exosomes are small vesicles containing proteins, nucleic acids, and biological lipids, which are responsible for intercellular communication. Studies have shown that exosomes can be utilized as effective drug delivery vehicles to accurately deliver therapeutic substances to target tissues, enhancing therapeutic effects and reducing side effects. Mesenchymal stem cells (MSCs) are a class of stem cells widely used for tissue engineering, regenerative medicine, and immunotherapy. Exosomes derived from MSCs have special immunomodulatory functions, low immunogenicity, the ability to penetrate tumor tissues, and high yield, which are expected to be engineered into efficient drug delivery systems. Despite the promising promise of MSC-derived exosomes, exploring their optimal preparation methods, drug-loading modalities, and therapeutic potential remains challenging. Therefore, this article reviews the related characteristics, preparation methods, application, and potential risks of MSC-derived exosomes as drug delivery systems in order to find potential therapeutic breakthroughs.
Collapse
Affiliation(s)
- Wenzhe Zhao
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Kaixuan Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Liangbo Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Ruichen Wang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Yang Lei
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Hui Yang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
| | - Leming Sun
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| |
Collapse
|
14
|
Li H, Zhang J, Tan M, Yin Y, Song Y, Zhao Y, Yan L, Li N, Zhang X, Bai J, Jiang T, Li H. Exosomes based strategies for cardiovascular diseases: Opportunities and challenges. Biomaterials 2024; 308:122544. [PMID: 38579591 DOI: 10.1016/j.biomaterials.2024.122544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024]
Abstract
Exosomes, as nanoscale extracellular vesicles (EVs), are secreted by all types of cells to facilitate intercellular communication in living organisms. After being taken up by neighboring or distant cells, exosomes can alter the expression levels of target genes in recipient cells and thereby affect their pathophysiological outcomes depending on payloads encapsulated therein. The functions and mechanisms of exosomes in cardiovascular diseases have attracted much attention in recent years and are thought to have cardioprotective and regenerative potential. This review summarizes the biogenesis and molecular contents of exosomes and details the roles played by exosomes released from various cells in the progression and recovery of cardiovascular disease. The review also discusses the current status of traditional exosomes in cardiovascular tissue engineering and regenerative medicine, pointing out several limitations in their application. It emphasizes that some of the existing emerging industrial or bioengineering technologies are promising to compensate for these shortcomings, and the combined application of exosomes and biomaterials provides an opportunity for mutual enhancement of their performance. The integration of exosome-based cell-free diagnostic and therapeutic options will contribute to the further development of cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Hang Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China; Department of Geriatrics, Cardiovascular Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yunfei Yin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Yiyi Song
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Yongjian Zhao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Lin Yan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China
| | - Ning Li
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, PR China
| | - Xianzuo Zhang
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, PR China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230022, PR China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, PR China.
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China.
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, PR China.
| |
Collapse
|
15
|
Liu M, Liu Y, Song T, Yang L, Qi L, Zhang YZ, Wang Y, Shen QT. Three-dimensional architecture of ESCRT-III flat spirals on the membrane. Proc Natl Acad Sci U S A 2024; 121:e2319115121. [PMID: 38709931 PMCID: PMC11098116 DOI: 10.1073/pnas.2319115121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/01/2024] [Indexed: 05/08/2024] Open
Abstract
The endosomal sorting complexes required for transport (ESCRTs) are responsible for membrane remodeling in many cellular processes, such as multivesicular body biogenesis, viral budding, and cytokinetic abscission. ESCRT-III, the most abundant ESCRT subunit, assembles into flat spirals as the primed state, essential to initiate membrane invagination. However, the three-dimensional architecture of ESCRT-III flat spirals remained vague for decades due to highly curved filaments with a small diameter and a single preferred orientation on the membrane. Here, we unveiled that yeast Snf7, a component of ESCRT-III, forms flat spirals on the lipid monolayers using cryogenic electron microscopy. We developed a geometry-constrained Euler angle-assigned reconstruction strategy and obtained moderate-resolution structures of Snf7 flat spirals with varying curvatures. Our analyses showed that Snf7 subunits recline on the membrane with N-terminal motifs α0 as anchors, adopt an open state with fused α2/3 helices, and bend α2/3 gradually from the outer to inner parts of flat spirals. In all, we provide the orientation and conformations of ESCRT-III flat spirals on the membrane and unveil the underlying assembly mechanism, which will serve as the initial step in understanding how ESCRTs drive membrane abscission.
Collapse
Affiliation(s)
- Mingdong Liu
- School of Life Sciences, Department of Chemical Biology, Southern University of Science and Technology, Shenzhen518055, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen518055, China
- iHuman Institute and School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
| | - Yunhui Liu
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen518055, China
| | - Tiefeng Song
- College of Life Sciences, Zhejiang University, Hangzhou310058, China
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, Haining314400, China
| | - Liuyan Yang
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao266237, China
| | - Lei Qi
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
- Biomedical Research Center for Structural Analysis, Shandong University, Jinan250012, China
| | - Yu-Zhong Zhang
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao266237, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou310058, China
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, Haining314400, China
| | - Qing-Tao Shen
- School of Life Sciences, Department of Chemical Biology, Southern University of Science and Technology, Shenzhen518055, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen518055, China
- iHuman Institute and School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
| |
Collapse
|
16
|
Fallahi S, Zangbar HS, Farajdokht F, Rahbarghazi R, Mohaddes G, Ghiasi F. Exosomes as a therapeutic tool to promote neurorestoration and cognitive function in neurological conditions: Achieve two ends with a single effort. CNS Neurosci Ther 2024; 30:e14752. [PMID: 38775149 PMCID: PMC11110007 DOI: 10.1111/cns.14752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/16/2024] [Accepted: 04/13/2024] [Indexed: 05/25/2024] Open
Abstract
Exosomes possess a significant role in intercellular communications. In the nervous system, various neural cells release exosomes that not only own a role in intercellular communications but also eliminate the waste of cells, maintain the myelin sheath, facilitate neurogenesis, and specifically assist in normal cognitive function. In neurological conditions including Parkinson's disease (PD), Alzheimer's disease (AD), traumatic brain injury (TBI), and stroke, exosomal cargo like miRNAs take part in the sequela of conditions and serve as a diagnostic tool of neurological disorders, too. Exosomes are not only a diagnostic tool but also their inhibition or administration from various sources like mesenchymal stem cells and serum, which have shown a worthy potential to treat multiple neurological disorders. In addition to neurodegenerative manifestations, cognitive deficiencies are an integral part of neurological diseases, and applying exosomes in improving both aspects of these diseases has been promising. This review discusses the status of exosome therapy in improving neurorestorative and cognitive function following neurological disease.
Collapse
Affiliation(s)
- Solmaz Fallahi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fereshteh Farajdokht
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Gisou Mohaddes
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Biomedical EducationCalifornia Health Sciences University, College of Osteopathic MedicineClovisCaliforniaUSA
| | - Fariba Ghiasi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
17
|
Zhou X, Jia Y, Mao C, Liu S. Small extracellular vesicles: Non-negligible vesicles in tumor progression, diagnosis, and therapy. Cancer Lett 2024; 580:216481. [PMID: 37972701 DOI: 10.1016/j.canlet.2023.216481] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
Small extracellular vesicles (sEVs) such as exosomes are nanoscale membranous particles (<200 nm) that have emerged as crucial targets for liquid biopsy and as promising drug delivery vehicles. They play a significant role in tumor progression as intercellular messengers. They can serve as biomarkers for tumor diagnosis and as drug carriers for cancer treatment. This article reviews recent studies on sEVs in oncology and explores their potential as biomarkers and drug delivery vehicles. Following tumorigenesis, sEVs in the tumor microenvironment (TME) and circulatory system undergo modifications to regulate various events in the TME, including angiogenesis, epithelial-mesenchymal transition (EMT), and tumor immunity, with either pro- or anti-tumor effects. sEVs have been investigated for use as diagnostic and prognostic biomarkers for a variety of tumors, including lung cancer, melanoma, breast cancer, prostate cancer, and hepatocellular carcinoma. sEVs can be used for cancer therapy by packaging drugs or proteins into them through pre- and post-isolation modification techniques. The clinical trials of sEVs as biomarkers and drug carriers are also summarized. Finally, the challenges in the use of sEVs are described and the possible approaches to tackling them are suggested. Overall, sEVs will advance the precision cancer medicine and has shown great potential in clinical applications.
Collapse
Affiliation(s)
- Xinru Zhou
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Yin Jia
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; School of Materials Science & Engineering, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shanrong Liu
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China.
| |
Collapse
|
18
|
Carlton JG, Baum B. Roles of ESCRT-III polymers in cell division across the tree of life. Curr Opin Cell Biol 2023; 85:102274. [PMID: 37944425 PMCID: PMC7615534 DOI: 10.1016/j.ceb.2023.102274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023]
Abstract
Every cell becomes two through a carefully orchestrated process of division. Prior to division, contractile machinery must first be assembled at the cell midzone to ensure that the cut, when it is made, bisects the two separated copies of the genetic material. Second, this contractile machinery must be dynamically tethered to the limiting plasma membrane so as to bring the membrane with it as it constricts. Finally, the connecting membrane must be severed to generate two physically separate daughter cells. In several organisms across the tree of life, Endosomal Sorting Complex Required for Transport (ESCRT)-III family proteins aid cell division by forming composite polymers that function together with the Vps4 AAA-ATPase to constrict and cut the membrane tube connecting nascent daughter cells from the inside. In this review, we discuss unique features of ESCRT-III that enable it to play this role in division in many archaea and eukaryotes.
Collapse
Affiliation(s)
- Jeremy Graham Carlton
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, SE1 1UL, UK; Organelle Dynamics Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Buzz Baum
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
19
|
Wang H, Gallet B, Moriscot C, Pezet M, Chatellard C, Kleman JP, Göttlinger H, Weissenhorn W, Boscheron C. An Inducible ESCRT-III Inhibition Tool to Control HIV-1 Budding. Viruses 2023; 15:2289. [PMID: 38140530 PMCID: PMC10748027 DOI: 10.3390/v15122289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
HIV-1 budding as well as many other cellular processes require the Endosomal Sorting Complex Required for Transport (ESCRT) machinery. Understanding the architecture of the native ESCRT-III complex at HIV-1 budding sites is limited due to spatial resolution and transient ESCRT-III recruitment. Here, we developed a drug-inducible transient HIV-1 budding inhibitory tool to enhance the ESCRT-III lifetime at budding sites. We generated autocleavable CHMP2A, CHMP3, and CHMP4B fusion proteins with the hepatitis C virus NS3 protease. We characterized the CHMP-NS3 fusion proteins in the absence and presence of protease inhibitor Glecaprevir with regard to expression, stability, localization, and HIV-1 Gag VLP budding. Immunoblotting experiments revealed rapid and stable accumulation of CHMP-NS3 fusion proteins. Notably, upon drug administration, CHMP2A-NS3 and CHMP4B-NS3 fusion proteins substantially decrease VLP release while CHMP3-NS3 exerted no effect but synergized with CHMP2A-NS3. Localization studies demonstrated the relocalization of CHMP-NS3 fusion proteins to the plasma membrane, endosomes, and Gag VLP budding sites. Through the combined use of transmission electron microscopy and video-microscopy, we unveiled drug-dependent accumulation of CHMP2A-NS3 and CHMP4B-NS3, causing a delay in HIV-1 Gag-VLP release. Our findings provide novel insight into the functional consequences of inhibiting ESCRT-III during HIV-1 budding and establish new tools to decipher the role of ESCRT-III at HIV-1 budding sites and other ESCRT-catalyzed cellular processes.
Collapse
Affiliation(s)
- Haiyan Wang
- University Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; (H.W.); (B.G.); (C.C.); (J.-P.K.)
| | - Benoit Gallet
- University Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; (H.W.); (B.G.); (C.C.); (J.-P.K.)
| | | | - Mylène Pezet
- University Grenoble Alpes, INSERM, IAB, 38000 Grenoble, France;
| | - Christine Chatellard
- University Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; (H.W.); (B.G.); (C.C.); (J.-P.K.)
| | - Jean-Philippe Kleman
- University Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; (H.W.); (B.G.); (C.C.); (J.-P.K.)
| | - Heinrich Göttlinger
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA;
| | - Winfried Weissenhorn
- University Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; (H.W.); (B.G.); (C.C.); (J.-P.K.)
| | - Cécile Boscheron
- University Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; (H.W.); (B.G.); (C.C.); (J.-P.K.)
| |
Collapse
|
20
|
Pfitzner AK, Zivkovic H, Bernat-Silvestre C, West M, Peltier T, Humbert F, Odorizzi G, Roux A. Vps60 initiates alternative ESCRT-III filaments. J Cell Biol 2023; 222:e202206028. [PMID: 37768378 PMCID: PMC10538557 DOI: 10.1083/jcb.202206028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 03/08/2023] [Accepted: 06/12/2023] [Indexed: 09/29/2023] Open
Abstract
Endosomal sorting complex required for transport-III (ESCRT-III) participates in essential cellular functions, from cell division to endosome maturation. The remarkable increase of its subunit diversity through evolution may have enabled the acquisition of novel functions. Here, we characterize a novel ESCRT-III copolymer initiated by Vps60. Membrane-bound Vps60 polymers recruit Vps2, Vps24, Did2, and Ist1, as previously shown for Snf7. Snf7- and Vps60-based filaments can coexist on membranes without interacting as their polymerization and recruitment of downstream subunits remain spatially and biochemically separated. In fibroblasts, Vps60/CHMP5 and Snf7/CHMP4 are both recruited during endosomal functions and cytokinesis, but their localization is segregated and their recruitment dynamics are different. Contrary to Snf7/CHMP4, Vps60/CHMP5 is not recruited during nuclear envelope reformation. Taken together, our results show that Vps60 and Snf7 form functionally distinct ESCRT-III polymers, supporting the notion that diversification of ESCRT-III subunits through evolution is linked to the acquisition of new cellular functions.
Collapse
Affiliation(s)
| | - Henry Zivkovic
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | | | - Matt West
- Department of Molecular Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Tanner Peltier
- Department of Molecular Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Frédéric Humbert
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Greg Odorizzi
- Department of Molecular Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
- National Center of Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
21
|
Hermosilla Aguayo V, Martin P, Tian N, Zheng J, Aho R, Losa M, Selleri L. ESCRT-dependent control of craniofacial morphogenesis with concomitant perturbation of NOTCH signaling. Dev Biol 2023; 503:25-42. [PMID: 37573008 DOI: 10.1016/j.ydbio.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Craniofacial development is orchestrated by transcription factor-driven regulatory networks, epigenetic modifications, and signaling pathways. Signaling molecules and their receptors rely on endo-lysosomal trafficking to prevent accumulation on the plasma membrane. ESCRT (Endosomal Sorting Complexes Required for Transport) machinery is recruited to endosomal membranes enabling degradation of such endosomal cargoes. Studies in vitro and in invertebrate models established the requirements of the ESCRT machinery in membrane remodeling, endosomal trafficking, and lysosomal degradation of activated membrane receptors. However, investigations during vertebrate development have been scarce. By ENU-induced mutagenesis, we isolated a mouse line, Vps25ENU/ENU, carrying a hypomorphic allele of the ESCRT-II component Vps25, with craniofacial anomalies resembling features of human congenital syndromes. Here, we assessed the spatiotemporal dynamics of Vps25 and additional ESCRT-encoding genes during murine development. We show that these genes are ubiquitously expressed although enriched in discrete domains of the craniofacial complex, heart, and limbs. ESCRT-encoding genes, including Vps25, are expressed in both cranial neural crest-derived mesenchyme and epithelium. Unlike constitutive ESCRT mutants, Vps25ENU/ENU embryos display late lethality. They exhibit hypoplastic lower jaw, stunted snout, dysmorphic ear pinnae, and secondary palate clefting. Thus, we provide the first evidence for critical roles of ESCRT-II in craniofacial morphogenesis and report perturbation of NOTCH signaling in craniofacial domains of Vps25ENU/ENU embryos. Given the known roles of NOTCH signaling in the developing cranium, and notably the lower jaw, we propose that the NOTCH pathway partly mediates the craniofacial defects of Vps25ENU/ENU mouse embryos.
Collapse
Affiliation(s)
- Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Peter Martin
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nuo Tian
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James Zheng
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robert Aho
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marta Losa
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
22
|
Schlösser L, Sachse C, Low HH, Schneider D. Conserved structures of ESCRT-III superfamily members across domains of life. Trends Biochem Sci 2023; 48:993-1004. [PMID: 37718229 DOI: 10.1016/j.tibs.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023]
Abstract
Structural and evolutionary studies of cyanobacterial phage shock protein A (PspA) and inner membrane-associated protein of 30 kDa (IM30) have revealed that these proteins belong to the endosomal sorting complex required for transport-III (ESCRT-III) superfamily, which is conserved across all three domains of life. PspA and IM30 share secondary and tertiary structures with eukaryotic ESCRT-III proteins, whilst also oligomerizing via conserved interactions. Here, we examine the structures of bacterial ESCRT-III-like proteins and compare the monomeric and oligomerized forms with their eukaryotic counterparts. We discuss conserved interactions used for self-assembly and highlight key hinge regions that mediate oligomer ultrastructure versatility. Finally, we address the differences in nomenclature assigned to equivalent structural motifs in both the bacterial and eukaryotic fields and suggest a common nomenclature applicable across the ESCRT-III superfamily.
Collapse
Affiliation(s)
- Lukas Schlösser
- Department of Chemistry, Biochemistry, Johannes Gutenberg University Mainz, Germany
| | - Carsten Sachse
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, ER-C-3/Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany; Institute for Biological Information Processing/IBI-6 Cellular Structural Biology, Jülich, Germany; Department of Biology, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Harry H Low
- Department of Infectious Disease, Imperial College, London, UK
| | - Dirk Schneider
- Department of Chemistry, Biochemistry, Johannes Gutenberg University Mainz, Germany; Institute of Molecular Physiology, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
23
|
Wang H, Gallet B, Moriscot C, Pezet M, Chatellard C, Kleman JP, Göttlinger H, Weissenhorn W, Boscheron C. An inducible ESCRT-III inhibition tool to control HIV-1 budding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562494. [PMID: 37905063 PMCID: PMC10614826 DOI: 10.1101/2023.10.16.562494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
HIV-1 budding as well as many other cellular processes require the Endosomal Sorting Complex Required for Transport (ESCRT) machinery. Understanding the architecture of the native ESCRT-III complex at HIV-1 budding sites is limited due to spatial resolution and transient ESCRT-III recruitment. Here, we developed a drug-inducible transient HIV-1 budding inhibitory tool to enhance the ESCRT-III lifetime at budding sites. We generated auto-cleavable CHMP2A, CHMP3, and CHMP4B fusion proteins with the hepatitis C virus NS3 protease. We characterized the CHMP-NS3 fusion proteins in the absence and presence of protease inhibitor Glecaprevir with regard to expression, stability, localization and HIV-1 Gag VLP budding. Immunoblotting experiments revealed rapid and stable accumulation of CHMP-NS3 fusion proteins with variable modification of Gag VLP budding upon drug administration. Notably, CHMP2A-NS3 and CHMP4B-NS3 fusion proteins substantially decrease VLP release while CHMP3-NS3 exerted a minor effect and synergized with CHMP2A-NS3. Localization studies demonstrated the re-localization of CHMP-NS3 fusion proteins to the plasma membrane, endosomes, and Gag VLP budding sites. Through the combined use of transmission electron microscopy and video-microscopy, we unveiled drug-dependent accumulation of CHMP2A-NS3 and CHMP4B-NS3, causing a delay in HIV-1 Gag-VLP release. Our findings provide novel insight into the functional consequences of inhibiting ESCRT-III during HIV-1 budding and establish new tools to decipher the role of ESCRT-III at HIV-1 budding sites and other ESCRT-catalyzed cellular processes.
Collapse
|
24
|
Ader NR, Chen L, Surovtsev IV, Chadwick WL, Rodriguez EC, King MC, Lusk CP. An ESCRT grommet cooperates with a diffusion barrier to maintain nuclear integrity. Nat Cell Biol 2023; 25:1465-1477. [PMID: 37783794 PMCID: PMC11365527 DOI: 10.1038/s41556-023-01235-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/17/2023] [Indexed: 10/04/2023]
Abstract
The molecular mechanisms by which the endosomal sorting complexes required for transport (ESCRT) proteins contribute to the integrity of the nuclear envelope (NE) barrier are not fully defined. We leveraged the single NE hole generated by mitotic extrusion of the Schizosaccharomyces pombe spindle pole body to reveal two modes of ESCRT function executed by distinct complements of ESCRT-III proteins, both dependent on CHMP7/Cmp7. A grommet-like function is required to restrict the NE hole in anaphase B, whereas replacement of Cmp7 by a sealing module ultimately closes the NE in interphase. Without Cmp7, nucleocytoplasmic compartmentalization remains intact despite NE discontinuities of up to 540 nm, suggesting mechanisms to limit diffusion through these holes. We implicate spindle pole body proteins as key components of a diffusion barrier acting with Cmp7 in anaphase B. Thus, NE remodelling mechanisms cooperate with proteinaceous diffusion barriers beyond nuclear pore complexes to maintain the nuclear compartment.
Collapse
Affiliation(s)
- Nicholas R Ader
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Linda Chen
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Ivan V Surovtsev
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
- Department of Physics, Yale University, New Haven, CT, USA
| | | | - Elisa C Rodriguez
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Megan C King
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA.
- Department of Molecular, Cell and Developmental Biology, Yale University, New Haven, CT, USA.
| | - C Patrick Lusk
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
25
|
Hurtig F, Burgers TC, Cezanne A, Jiang X, Mol FN, Traparić J, Pulschen AA, Nierhaus T, Tarrason-Risa G, Harker-Kirschneck L, Löwe J, Šarić A, Vlijm R, Baum B. The patterned assembly and stepwise Vps4-mediated disassembly of composite ESCRT-III polymers drives archaeal cell division. SCIENCE ADVANCES 2023; 9:eade5224. [PMID: 36921039 PMCID: PMC10017037 DOI: 10.1126/sciadv.ade5224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/14/2023] [Indexed: 05/13/2023]
Abstract
ESCRT-III family proteins form composite polymers that deform and cut membrane tubes in the context of a wide range of cell biological processes across the tree of life. In reconstituted systems, sequential changes in the composition of ESCRT-III polymers induced by the AAA-adenosine triphosphatase Vps4 have been shown to remodel membranes. However, it is not known how composite ESCRT-III polymers are organized and remodeled in space and time in a cellular context. Taking advantage of the relative simplicity of the ESCRT-III-dependent division system in Sulfolobus acidocaldarius, one of the closest experimentally tractable prokaryotic relatives of eukaryotes, we use super-resolution microscopy, electron microscopy, and computational modeling to show how CdvB/CdvB1/CdvB2 proteins form a precisely patterned composite ESCRT-III division ring, which undergoes stepwise Vps4-dependent disassembly and contracts to cut cells into two. These observations lead us to suggest sequential changes in a patterned composite polymer as a general mechanism of ESCRT-III-dependent membrane remodeling.
Collapse
Affiliation(s)
- Fredrik Hurtig
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Thomas C. Q. Burgers
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, Netherlands
| | - Alice Cezanne
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Xiuyun Jiang
- Laboratory of Soft Matter Physics, The Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Frank N. Mol
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, Netherlands
| | - Jovan Traparić
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Tim Nierhaus
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Lena Harker-Kirschneck
- University College London, Institute for the Physics of Living Systems, WC1E 6BT London, UK
| | - Jan Löwe
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Anđela Šarić
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Rifka Vlijm
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, Netherlands
| | - Buzz Baum
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| |
Collapse
|
26
|
Jukic N, Perrino AP, Redondo-Morata L, Scheuring S. Structure and dynamics of ESCRT-III membrane remodeling proteins by high-speed atomic force microscopy. J Biol Chem 2023; 299:104575. [PMID: 36870686 PMCID: PMC10074808 DOI: 10.1016/j.jbc.2023.104575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Endosomal Sorting Complex Required for Transport (ESCRT) proteins assemble on the cytoplasmic leaflet of membranes and remodel them. ESCRT is involved in biological processes where membranes are bent away from the cytosol, constricted, and finally severed, such as in multi-vesicular body formation (in the endosomal pathway for protein sorting) or abscission during cell division. The ESCRT system is hijacked by enveloped viruses to allow buds of nascent virions to be constricted, severed and released. ESCRT-III proteins, the most downstream components of the ESCRT system, are monomeric and cytosolic in their autoinhibited conformation. They share a common architecture, a four-helix bundle with a fifth helix that interacts with this bundle to prevent polymerizing. Upon binding to negatively charged membranes, the ESCRT-III components adopt an activated state that allows them to polymerize into filaments and spirals, and to interact with the AAA-ATPase Vps4 for polymer remodeling. ESCRT-III has been studied with electron microscopy (EM) and fluorescence microscopy (FM); these methods provided invaluable information about ESCRT assembly structures or their dynamics, respectively, but neither approach provides detailed insights into both aspects simultaneously. High-speed atomic force microscopy (HS-AFM) has overcome this shortcoming, providing movies at high spatio-temporal resolution of biomolecular processes, significantly increasing our understanding of ESCRT-III structure and dynamics. Here, we review the contributions of HS-AFM in the analysis of ESCRT-III, focusing on recent developments of non-planar and deformable HS-AFM supports. We divide the HS-AFM observations into four sequential steps in the ESCRT-III lifecycle: 1) polymerization, 2) morphology, 3) dynamics, and 4) depolymerization.
Collapse
Affiliation(s)
- Nebojsa Jukic
- Weill Cornell Medicine, Physiology, Biophysics and Systems Biology Graduate Program, New York, NY 10065, USA
| | - Alma P Perrino
- Weill Cornell Medicine, Department of Anesthesiology, 1300 York Avenue, New York, NY 10065, USA
| | - Lorena Redondo-Morata
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Simon Scheuring
- Weill Cornell Medicine, Department of Anesthesiology, 1300 York Avenue, New York, NY 10065, USA; Weill Cornell Medicine, Department of Physiology and Biophysics, 1300 York Avenue, New York, NY 10065, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, NY 14853, USA.
| |
Collapse
|
27
|
Marie PP, Fan S, Mason J, Wells A, Mendes CC, Wainwright SM, Scott S, Fischer R, Harris AL, Wilson C, Goberdhan DCI. Accessory ESCRT-III proteins are conserved and selective regulators of Rab11a-exosome formation. J Extracell Vesicles 2023; 12:e12311. [PMID: 36872252 PMCID: PMC9986085 DOI: 10.1002/jev2.12311] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 01/18/2023] [Accepted: 02/09/2023] [Indexed: 03/07/2023] Open
Abstract
Exosomes are secreted nanovesicles with potent signalling activity that are initially formed as intraluminal vesicles (ILVs) in late Rab7-positive multivesicular endosomes, and also in recycling Rab11a-positive endosomes, particularly under some forms of nutrient stress. The core proteins of the Endosomal Sorting Complex Required for Transport (ESCRT) participate in exosome biogenesis and ILV-mediated destruction of ubiquitinylated cargos. Accessory ESCRT-III components have reported roles in ESCRT-III-mediated vesicle scission, but their precise functions are poorly defined. They frequently only appear essential under stress. Comparative proteomics analysis of human small extracellular vesicles revealed that accessory ESCRT-III proteins, CHMP1A, CHMP1B, CHMP5 and IST1, are increased in Rab11a-enriched exosome preparations. We show that these proteins are required to form ILVs in Drosophila secondary cell recycling endosomes, but unlike core ESCRTs, they are not involved in degradation of ubiquitinylated proteins in late endosomes. Furthermore, CHMP5 knockdown in human HCT116 colorectal cancer cells selectively inhibits Rab11a-exosome production. Accessory ESCRT-III knockdown suppresses seminal fluid-mediated reproductive signalling by secondary cells and the growth-promoting activity of Rab11a-exosome-containing EVs from HCT116 cells. We conclude that accessory ESCRT-III components have a specific, ubiquitin-independent role in Rab11a-exosome generation, a mechanism that might be targeted to selectively block pro-tumorigenic activities of these vesicles in cancer.
Collapse
Affiliation(s)
- Pauline P. Marie
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Shih‐Jung Fan
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - John Mason
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Adam Wells
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Cláudia C. Mendes
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - S. Mark Wainwright
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Sheherezade Scott
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Roman Fischer
- Target Discovery InstituteUniversity of OxfordOxfordUK
| | | | - Clive Wilson
- Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | | |
Collapse
|
28
|
Romano JD, Mayoral J, Guevara RB, Rivera-Cuevas Y, Carruthers VB, Weiss LM, Coppens I. Toxoplasma gondii scavenges mammalian host organelles through the usurpation of host ESCRT-III and Vps4A. J Cell Sci 2023; 136:jcs260159. [PMID: 36718630 PMCID: PMC10022688 DOI: 10.1242/jcs.260159] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Intracellular pathogens exploit cellular resources through host cell manipulation. Within its nonfusogenic parasitophorous vacuole (PV), Toxoplasma gondii targets host nutrient-filled organelles and sequesters them into the PV through deep invaginations of the PV membrane (PVM) that ultimately detach from this membrane. Some of these invaginations are generated by an intravacuolar network (IVN) of parasite-derived tubules attached to the PVM. Here, we examined the usurpation of host ESCRT-III and Vps4A by the parasite to create PVM buds and vesicles. CHMP4B associated with the PVM/IVN, and dominant-negative (DN) CHMP4B formed many long PVM invaginations containing CHMP4B filaments. These invaginations were shorter in IVN-deficient parasites, suggesting cooperation between the IVN and ESCRT. In infected cells expressing Vps4A-DN, enlarged intra-PV structures containing host endolysosomes accumulated, reflecting defects in PVM scission. Parasite mutants lacking T. gondii (Tg)GRA14 or TgGRA64, which interact with ESCRT, reduced CHMP4B-DN-induced PVM invaginations and intra-PV host organelles, with greater defects in a double knockout, revealing the exploitation of ESCRT to scavenge host organelles by Toxoplasma.
Collapse
Affiliation(s)
- Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Joshua Mayoral
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rebekah B. Guevara
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yolanda Rivera-Cuevas
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Vern B. Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
29
|
Azad K, Guilligay D, Boscheron C, Maity S, De Franceschi N, Sulbaran G, Effantin G, Wang H, Kleman JP, Bassereau P, Schoehn G, Roos WH, Desfosses A, Weissenhorn W. Structural basis of CHMP2A-CHMP3 ESCRT-III polymer assembly and membrane cleavage. Nat Struct Mol Biol 2023; 30:81-90. [PMID: 36604498 DOI: 10.1038/s41594-022-00867-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 10/12/2022] [Indexed: 01/07/2023]
Abstract
The endosomal sorting complex required for transport (ESCRT) is a highly conserved protein machinery that drives a divers set of physiological and pathological membrane remodeling processes. However, the structural basis of ESCRT-III polymers stabilizing, constricting and cleaving negatively curved membranes is yet unknown. Here we present cryo-EM structures of membrane-coated CHMP2A-CHMP3 filaments from Homo sapiens of two different diameters at 3.3 and 3.6 Å resolution. The structures reveal helical filaments assembled by CHMP2A-CHMP3 heterodimers in the open ESCRT-III conformation, which generates a partially positive charged membrane interaction surface, positions short N-terminal motifs for membrane interaction and the C-terminal VPS4 target sequence toward the tube interior. Inter-filament interactions are electrostatic, which may facilitate filament sliding upon VPS4-mediated polymer remodeling. Fluorescence microscopy as well as high-speed atomic force microscopy imaging corroborate that VPS4 can constrict and cleave CHMP2A-CHMP3 membrane tubes. We therefore conclude that CHMP2A-CHMP3-VPS4 act as a minimal membrane fission machinery.
Collapse
Affiliation(s)
- Kimi Azad
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Delphine Guilligay
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Cecile Boscheron
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Sourav Maity
- Moleculaire Biofysica, Zernike Institute, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Nicola De Franceschi
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France.,Curie Institute, Laboratory of Physical Chemistry Curie, University of PSL, Sorbonne University, CNRS, Paris, France
| | - Guidenn Sulbaran
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Gregory Effantin
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Haiyan Wang
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Jean-Philippe Kleman
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Patricia Bassereau
- Curie Institute, Laboratory of Physical Chemistry Curie, University of PSL, Sorbonne University, CNRS, Paris, France
| | - Guy Schoehn
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Institute, Rijksuniversiteit Groningen, Groningen, the Netherlands
| | - Ambroise Desfosses
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France.
| | - Winfried Weissenhorn
- Institute of Structural Biology (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France.
| |
Collapse
|
30
|
Nachmias D, Melnikov N, Zorea A, Sharon M, Yemini R, De-Picchoto Y, Tsirkas I, Aharoni A, Frohn B, Schwille P, Zarivach R, Mizrahi I, Elia N. Asgard ESCRT-III and VPS4 reveal conserved chromatin binding properties of the ESCRT machinery. THE ISME JOURNAL 2023; 17:117-129. [PMID: 36221007 PMCID: PMC9751279 DOI: 10.1038/s41396-022-01328-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
The archaeal Asgard superphylum currently stands as the most promising prokaryotic candidate, from which eukaryotic cells emerged. This unique superphylum encodes for eukaryotic signature proteins (ESP) that could shed light on the origin of eukaryotes, but the properties and function of these proteins is largely unresolved. Here, we set to understand the function of an Asgard archaeal protein family, namely the ESCRT machinery, that is conserved across all domains of life and executes basic cellular eukaryotic functions, including membrane constriction during cell division. We find that ESCRT proteins encoded in Loki archaea, express in mammalian and yeast cells, and that the Loki ESCRT-III protein, CHMP4-7, resides in the eukaryotic nucleus in both organisms. Moreover, Loki ESCRT-III proteins associated with chromatin, recruited their AAA-ATPase VPS4 counterpart to organize in discrete foci in the mammalian nucleus, and directly bind DNA. The human ESCRT-III protein, CHMP1B, exhibited similar nuclear properties and recruited both human and Asgard VPS4s to nuclear foci, indicating interspecies interactions. Mutation analysis revealed a role for the N terminal region of ESCRT-III in mediating these phenotypes in both human and Asgard ESCRTs. These findings suggest that ESCRT proteins hold chromatin binding properties that were highly preserved through the billion years of evolution separating Asgard archaea and humans. The conserved chromatin binding properties of the ESCRT membrane remodeling machinery, reported here, may have important implications for the origin of eukaryogenesis.
Collapse
Affiliation(s)
- Dikla Nachmias
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Nataly Melnikov
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Alvah Zorea
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Maya Sharon
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Reut Yemini
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Yasmin De-Picchoto
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Ioannis Tsirkas
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Amir Aharoni
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Bela Frohn
- Department of Cellular and Molecular Biophysics, Max-Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Petra Schwille
- Department of Cellular and Molecular Biophysics, Max-Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Raz Zarivach
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Itzhak Mizrahi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Natalie Elia
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
| |
Collapse
|
31
|
Meadowcroft B, Palaia I, Pfitzner AK, Roux A, Baum B, Šarić A. Mechanochemical Rules for Shape-Shifting Filaments that Remodel Membranes. PHYSICAL REVIEW LETTERS 2022; 129:268101. [PMID: 36608212 DOI: 10.1103/physrevlett.129.268101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
The sequential exchange of filament composition to increase filament curvature was proposed as a mechanism for how some biological polymers deform and cut membranes. The relationship between the filament composition and its mechanical effect is lacking. We develop a kinetic model for the assembly of composite filaments that includes protein-membrane adhesion, filament mechanics and membrane mechanics. We identify the physical conditions for such a membrane remodeling and show this mechanism of sequential polymer assembly lowers the energetic barrier for membrane deformation.
Collapse
Affiliation(s)
- Billie Meadowcroft
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Ivan Palaia
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | | | - Aurélien Roux
- Biochemistry Department, University of Geneva, CH-1211 Geneva, Switzerland
- Swiss National Centre for Competence in Research Programme Chemical Biology, CH-1211 Geneva, Switzerland
| | - Buzz Baum
- MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Anđela Šarić
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| |
Collapse
|
32
|
Avalos-Padilla Y, Georgiev VN, Ewins E, Robinson T, Orozco E, Lipowsky R, Dimova R. Stepwise remodeling and subcompartment formation in individual vesicles by three ESCRT-III proteins. iScience 2022; 26:105765. [PMID: 36590172 PMCID: PMC9800321 DOI: 10.1016/j.isci.2022.105765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/21/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) is a multi-protein machinery involved in several membrane remodeling processes. Different approaches have been used to resolve how ESCRT proteins scission membranes. However, the underlying mechanisms generating membrane deformations are still a matter of debate. Here, giant unilamellar vesicles, microfluidic technology, and micropipette aspiration are combined to continuously follow the ESCRT-III-mediated membrane remodeling on the single-vesicle level for the first time. With this approach, we identify different mechanisms by which a minimal set of three ESCRT-III proteins from Entamoeba histolytica reshape the membrane. These proteins modulate the membrane stiffness and spontaneous curvature to regulate bud size and generate intraluminal vesicles even in the absence of ATP. We demonstrate that the bud stability depends on the protein concentration and membrane tension. The approaches introduced here should open the road to diverse applications in synthetic biology for establishing artificial cells with several membrane compartments.
Collapse
Affiliation(s)
- Yunuen Avalos-Padilla
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14476 Potsdam, Germany,Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, ES-08028 Barcelona, Spain,Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain
| | - Vasil N. Georgiev
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14476 Potsdam, Germany
| | - Eleanor Ewins
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14476 Potsdam, Germany
| | - Tom Robinson
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14476 Potsdam, Germany
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV IPN, 07360 Ciudad de México, México
| | - Reinhard Lipowsky
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14476 Potsdam, Germany
| | - Rumiana Dimova
- Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14476 Potsdam, Germany,Corresponding author
| |
Collapse
|
33
|
Hussen BM, Faraj GSH, Rasul MF, Hidayat HJ, Salihi A, Baniahmad A, Taheri M, Ghafouri-Frad S. Strategies to overcome the main challenges of the use of exosomes as drug carrier for cancer therapy. Cancer Cell Int 2022; 22:323. [PMID: 36258195 PMCID: PMC9580186 DOI: 10.1186/s12935-022-02743-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
Exosomes are naturally occurring nanosized particles that aid intercellular communication by transmitting biological information between cells. Exosomes have therapeutic efficacy that can transfer their contents between cells as natural carriers. In addition, the exosomal contents delivered to the recipient pathological cells significantly inhibit cancer progression. However, exosome-based tumor treatments are inadequately precise or successful, and various challenges should be adequately overcome. Here, we discuss the significant challenges that exosomes face as drug carriers used for therapeutic targets and strategies for overcoming these challenges in order to promote this new incoming drug carrier further and improve future clinical outcomes. We also present techniques for overcoming these challenges.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Goran Sedeeq Hama Faraj
- College of Medicine, Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaymaniyah, Iraq
| | - Mohammad Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University, Erbil, Kurdistan Region, Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University, Erbil, Kurdistan Region, Iraq
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany. .,Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Frad
- Department of Medical Genetics,, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Jiang X, Harker-Kirschneck L, Vanhille-Campos C, Pfitzner AK, Lominadze E, Roux A, Baum B, Šarić A. Modelling membrane reshaping by staged polymerization of ESCRT-III filaments. PLoS Comput Biol 2022; 18:e1010586. [PMID: 36251703 PMCID: PMC9612822 DOI: 10.1371/journal.pcbi.1010586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/27/2022] [Accepted: 09/19/2022] [Indexed: 12/24/2022] Open
Abstract
ESCRT-III filaments are composite cytoskeletal polymers that can constrict and cut cell membranes from the inside of the membrane neck. Membrane-bound ESCRT-III filaments undergo a series of dramatic composition and geometry changes in the presence of an ATP-consuming Vps4 enzyme, which causes stepwise changes in the membrane morphology. We set out to understand the physical mechanisms involved in translating the changes in ESCRT-III polymer composition into membrane deformation. We have built a coarse-grained model in which ESCRT-III polymers of different geometries and mechanical properties are allowed to copolymerise and bind to a deformable membrane. By modelling ATP-driven stepwise depolymerisation of specific polymers, we identify mechanical regimes in which changes in filament composition trigger the associated membrane transition from a flat to a buckled state, and then to a tubule state that eventually undergoes scission to release a small cargo-loaded vesicle. We then characterise how the location and kinetics of polymer loss affects the extent of membrane deformation and the efficiency of membrane neck scission. Our results identify the near-minimal mechanical conditions for the operation of shape-shifting composite polymers that sever membrane necks.
Collapse
Affiliation(s)
- Xiuyun Jiang
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Lena Harker-Kirschneck
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Christian Vanhille-Campos
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Elene Lominadze
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, United Kingdom
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Buzz Baum
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Anđela Šarić
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
35
|
Polymyxin B Reduces Brain Injury in Ischemic Stroke Rat Through a Mechanism Involving Targeting ESCRT-III Machinery and RIPK1/RIPK3/MLKL Pathway. J Cardiovasc Transl Res 2022; 15:1129-1142. [PMID: 35239171 DOI: 10.1007/s12265-022-10224-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/21/2022] [Indexed: 02/05/2023]
Abstract
Endosomal sorting complex required for transport III (ESCRT-III) machinery is a key component to counteract the mixed lineage kinase domain-like pseudokinase (MLKL)-induced plasma membrane broken in cells undergoing necroptosis. Based on the bioinformatics analysis, polymyxin B, a polypeptide antibiotic, is predicted to simultaneously interact with ESCRT-III subunits and necroptosis-relevant proteins. This study aims to explore whether polymyxin B could reduce necroptosis in the stroke rat brain via enhancing the ESCRT-III machinery and/or suppressing the RIPK1/RIPK3/MLKL pathway. The stroke rats showed evident brain injury, concomitant with the downregulation of ESCRT-III subunits and the upregulation of necroptosis-relevant proteins. Post-ischemic administration of polymyxin B could alleviate the brain injury, accompanied by restoration of the levels of ESCRT-III subunits and suppression of necroptosis-relevant proteins. And, polymyxin B exerted similar effects in hypoxia-treated HT22 cells. We conclude that polymyxin B can reduce necroptosis in the stroke rat brain via enhancing the ESCRT-III machinery and suppressing the RIPK1/RIPK3/MLKL pathway simultaneously.
Collapse
|
36
|
Arines FM, Li M. An optimized protocol to analyze membrane protein degradation in yeast using quantitative western blot and flow cytometry. STAR Protoc 2022; 3:101274. [PMID: 35403002 PMCID: PMC8987394 DOI: 10.1016/j.xpro.2022.101274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2022] Open
Abstract
Membrane proteins (MPs) are essential in many cellular functions. To maintain proteostasis, MPs are downregulated via ubiquitination and degradation. Here, we describe an optimized protocol to analyze MP degradation using quantitative western blot and flow cytometry-based approaches. We use the degradation of Ypq1, a vacuole membrane lysine transporter, to demonstrate the protocol, which can be adapted for other organelle MPs and thus provide useful tools to study MP regulation in yeast and other model organisms. For complete details on the use and execution of this protocol, please refer to Arines et al. (2021) and Yang et al. (2020). Two complementary protocols for quantifying membrane protein degradation in yeast Measures the decrease of full-length protein by quantitative western blot Quantifies the quenching of GFP fluorescence by flow cytometry Adaptable to other fluorescent protein tags and mammalian cells
Collapse
|
37
|
Jukic N, Perrino AP, Humbert F, Roux A, Scheuring S. Snf7 spirals sense and alter membrane curvature. Nat Commun 2022; 13:2174. [PMID: 35449207 PMCID: PMC9023468 DOI: 10.1038/s41467-022-29850-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/03/2022] [Indexed: 11/25/2022] Open
Abstract
Endosomal Sorting Complex Required for Transport III (ESCRT-III) is a conserved protein system involved in many cellular processes resulting in membrane deformation and scission, topologically away from the cytoplasm. However, little is known about the transition of the planar membrane-associated protein assembly into a 3D structure. High-speed atomic force microscopy (HS-AFM) provided insights into assembly, structural dynamics and turnover of Snf7, the major ESCRT-III component, on planar supported lipid bilayers. Here, we develop HS-AFM experiments that remove the constraints of membrane planarity, crowdedness, and support rigidity. On non-planar membranes, Snf7 monomers are curvature insensitive, but Snf7-spirals selectively adapt their conformation to membrane geometry. In a non-crowded system, Snf7-spirals reach a critical radius, and remodel to minimize internal stress. On non-rigid supports, Snf7-spirals compact and buckle, deforming the underlying bilayer. These experiments provide direct evidence that Snf7 is sufficient to mediate topological transitions, in agreement with the loaded spiral spring model.
Collapse
Affiliation(s)
- Nebojsa Jukic
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Alma P Perrino
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Frédéric Humbert
- Department of Biochemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, CH-1211, Geneva, Switzerland
- Swiss National Centre for Competence in Research Programme Chemical Biology, CH-1211, Geneva, Switzerland
| | - Simon Scheuring
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, NY, 14853, USA.
| |
Collapse
|
38
|
Banjade S, Zhu L, Jorgensen JR, Suzuki SW, Emr SD. Recruitment and organization of ESCRT-0 and ubiquitinated cargo via condensation. SCIENCE ADVANCES 2022; 8:eabm5149. [PMID: 35363519 PMCID: PMC10938570 DOI: 10.1126/sciadv.abm5149] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
The general mechanisms by which ESCRTs (Endosomal Sorting Complexes Required for Transport) are specifically recruited to various membranes, and how ESCRT subunits are spatially organized remain central questions in cell biology. At the endosome and lysosomes, ubiquitination of membrane proteins triggers ESCRT-mediated substrate recognition and degradation. Using the yeast lysosome/vacuole, we define the principles by which substrate engagement by ESCRTs occurs at this organelle. We find that multivalent interactions between ESCRT-0 and polyubiquitin are critical for substrate recognition at yeast vacuoles, with a lower-valency requirement for cargo engagement at endosomes. Direct recruitment of ESCRT-0 induces dynamic foci on the vacuole membrane and forms fluid condensates in vitro with polyubiquitin. We propose that self-assembly of early ESCRTs induces condensation, an initial step in ESCRT assembly/nucleation at membranes. This property can be tuned specifically at various organelles by modulating the number of binding interactions.
Collapse
Affiliation(s)
- Sudeep Banjade
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Lu Zhu
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Jeffrey R. Jorgensen
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Sho W. Suzuki
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Scott D. Emr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
39
|
Li Y, Huang L, Chen Y, Shi Y, Ze Y, Yao Y. Irradiated cell-derived exosomes transmit essential molecules inducing radiotherapy resistance. Int J Radiat Oncol Biol Phys 2022; 113:192-202. [PMID: 35217095 DOI: 10.1016/j.ijrobp.2022.01.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/11/2021] [Accepted: 01/23/2022] [Indexed: 02/05/2023]
Abstract
Radio-resistance has always been a major obstacle in radiation therapy (RT) progress. Radiotherapy (RT) leads to changes in the contents of released exosomes. The researches have shown that irradiated cell-derived exosomes influence recipient cell proliferation, migration, cell cycle arrest and apoptosis. All evidence indicates that exosomes play a significant role in radio-resistance. In this review, we describe the potential role of exosomes in cancer. We summarize that the irradiated cell-derived exosomes influence radio-resistance in recipient cells by three main mechanisms: 1) enhancing DNA repair, 2) regulating cell death signalling pathways, 3) inducing cancer cells to exhibit stem cell properties. We also discuss that the origin of the phenomenon might be the changes of molecular mechanisms of irradiated cell-derived exosomes formation affected by RT. Further, targeting exosomes as an adjuvant therapy might be a promising way for cancer treatments.
Collapse
Affiliation(s)
- Yiling Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Linyang Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yanchi Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yixin Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yiting Ze
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
40
|
Harker-Kirschneck L, Hafner AE, Yao T, Vanhille-Campos C, Jiang X, Pulschen A, Hurtig F, Hryniuk D, Culley S, Henriques R, Baum B, Šarić A. Physical mechanisms of ESCRT-III-driven cell division. Proc Natl Acad Sci U S A 2022; 119:e2107763119. [PMID: 34983838 PMCID: PMC8740586 DOI: 10.1073/pnas.2107763119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2021] [Indexed: 11/25/2022] Open
Abstract
Living systems propagate by undergoing rounds of cell growth and division. Cell division is at heart a physical process that requires mechanical forces, usually exerted by assemblies of cytoskeletal polymers. Here we developed a physical model for the ESCRT-III-mediated division of archaeal cells, which despite their structural simplicity share machinery and evolutionary origins with eukaryotes. By comparing the dynamics of simulations with data collected from live cell imaging experiments, we propose that this branch of life uses a previously unidentified division mechanism. Active changes in the curvature of elastic cytoskeletal filaments can lead to filament perversions and supercoiling, to drive ring constriction and deform the overlying membrane. Abscission is then completed following filament disassembly. The model was also used to explore how different adenosine triphosphate (ATP)-driven processes that govern the way the structure of the filament is changed likely impact the robustness and symmetry of the resulting division. Comparisons between midcell constriction dynamics in simulations and experiments reveal a good agreement with the process when changes in curvature are implemented at random positions along the filament, supporting this as a possible mechanism of ESCRT-III-dependent division in this system. Beyond archaea, this study pinpoints a general mechanism of cytokinesis based on dynamic coupling between a coiling filament and the membrane.
Collapse
Affiliation(s)
- Lena Harker-Kirschneck
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Anne E Hafner
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Tina Yao
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
| | - Christian Vanhille-Campos
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Xiuyun Jiang
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Andre Pulschen
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - Fredrik Hurtig
- Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - Dawid Hryniuk
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
| | - Siân Culley
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Ricardo Henriques
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Buzz Baum
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, United Kingdom
| | - Anđela Šarić
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom;
- Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
41
|
Parkinson G, Roboti P, Zhang L, Taylor S, Woodman P. His domain protein tyrosine phosphatase and Rabaptin-5 couple endo-lysosomal sorting of EGFR with endosomal maturation. J Cell Sci 2021; 134:272512. [PMID: 34657963 PMCID: PMC8627557 DOI: 10.1242/jcs.259192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/13/2021] [Indexed: 01/20/2023] Open
Abstract
His domain protein tyrosine phosphatase (HD-PTP; also known as PTPN23) collaborates with endosomal sorting complexes required for transport (ESCRTs) to sort endosomal cargo into intralumenal vesicles, forming the multivesicular body (MVB). Completion of MVB sorting is accompanied by maturation of the endosome into a late endosome, an event that requires inactivation of the early endosomal GTPase Rab5 (herein referring to generically to all isoforms). Here, we show that HD-PTP links ESCRT function with endosomal maturation. HD-PTP depletion prevents MVB sorting, while also blocking cargo from exiting Rab5-rich endosomes. HD-PTP-depleted cells contain hyperphosphorylated Rabaptin-5 (also known as RABEP1), a cofactor for the Rab5 guanine nucleotide exchange factor Rabex-5 (also known as RABGEF1), although HD-PTP is unlikely to directly dephosphorylate Rabaptin-5. In addition, HD-PTP-depleted cells exhibit Rabaptin-5-dependent hyperactivation of Rab5. HD-PTP binds directly to Rabaptin-5, between its Rabex-5- and Rab5-binding domains. This binding reaction involves the ESCRT-0/ESCRT-III binding site in HD-PTP, which is competed for by an ESCRT-III peptide. Jointly, these findings indicate that HD-PTP may alternatively scaffold ESCRTs and modulate Rabex-5–Rabaptin-5 activity, thereby helping to coordinate the completion of MVB sorting with endosomal maturation. Summary: Sorting of endocytic cargo to the multivesicular body is accompanied by endosomal maturation. Here, we provide a potential mechanism by which these two processes are linked.
Collapse
Affiliation(s)
- Gabrielle Parkinson
- Faculty of Biology, Medicine and Health, Manchester Academic and Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Peristera Roboti
- Faculty of Biology, Medicine and Health, Manchester Academic and Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Ling Zhang
- Faculty of Biology, Medicine and Health, Manchester Academic and Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Sandra Taylor
- Faculty of Biology, Medicine and Health, Manchester Academic and Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Philip Woodman
- Faculty of Biology, Medicine and Health, Manchester Academic and Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
42
|
ESCRT-III induces phase separation in model membranes prior to budding and causes invagination of the liquid-ordered phase. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183689. [PMID: 34224704 DOI: 10.1016/j.bbamem.2021.183689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/05/2021] [Accepted: 06/27/2021] [Indexed: 11/21/2022]
Abstract
Membrane fission triggered by the endosomal sorting complex required for transport (ESCRT) is an important process observed in several pathogenic and non-pathogenic cellular events. From a synthetic-biology viewpoint, ESCRT proteins represent an interesting machinery for the construction of cell mimetic sub-compartments produced by fission. Since their discovery, the studies on ESCRT-III-mediated action, have mainly focused on protein dynamics, ignoring the role of lipid organization and membrane phase state. Recently, it has been suggested that membrane buds formed by the action of ESCRT-III are generated from transient microdomains in endosomal membranes. However, the interplay between membrane domain formation and ESCRT remodeling pathways has not been investigated. Here, giant unilamellar vesicles made of ternary lipid mixtures, either homogeneous in phase or exhibiting liquid-ordered/liquid-disordered phase coexistence, were employed as a model membrane system. These vesicles were incubated with purified recombinant ESCRT-III proteins from the parasite Entamoeba histolytica. In homogeneous membranes, we observe that EhVps32 can trigger domain formation while EhVps20 preferentially co-localizes in the liquid disordered phase. The addition of EhVps24 appears to induce the formation of intraluminal vesicles produced from the liquid-ordered phase. In phase separated membranes, the intraluminal vesicles are also generated from the liquid-ordered phase and presumably emerge from the phase boundary region. Our findings reinforce the hypothesis that ESCRT-mediated remodeling depends on the membrane phase state. Furthermore, the obtained results point to a potential synthetic biology approach for establishing eukaryotic mimics of artificial cells with microcompartments of specific membrane composition, which can also differ from that of the mother vesicle.
Collapse
|
43
|
Liu C, Zeng Y, Li H, Yang C, Shen W, Xu M, Xiao Z, Chen T, Li B, Cao W, Jiang L, Otegui MS, Gao C. A plant-unique ESCRT component, FYVE4, regulates multivesicular endosome biogenesis and plant growth. THE NEW PHYTOLOGIST 2021; 231:193-209. [PMID: 33772801 DOI: 10.1111/nph.17358] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/18/2021] [Indexed: 06/12/2023]
Abstract
During evolution, land plants generated unique proteins that participate in endosomal sorting and multivesicular endosome (MVE) biogenesis, many of them with specific phosphoinositide-binding capabilities. Nonetheless, the function of most plant phosphoinositide-binding proteins in endosomal trafficking remains elusive. Here, we analysed several Arabidopsis mutants lacking predicted phosphoinositide-binding proteins and first identified fyve4-1 as a mutant with a hypersensitive response to high-boron conditions and defects in degradative vacuolar sorting of membrane proteins such as the borate exporter BOR1-GFP. FYVE4 encodes a plant-unique, FYVE domain-containing protein that interacts with SNF7, a core component of ESCRT-III (Endosomal Sorting Complex Required for Transport III). FYVE4 affects the membrane association of the late-acting ESCRT components SNF7 and VPS4, and modulates the formation of intraluminal vesicles (ILVs) inside MVEs. The critical function of FYVE4 in the ESCRT pathway was further demonstrated by the strong genetic interactions with SNF7B and LIP5. Although the fyve4-1, snf7b and lip5 single mutants were viable, the fyve4-1 snf7b and fyve4-1 lip5 double mutants were seedling lethal, with strong defects in MVE biogenesis and vacuolar sorting of ubiquitinated membrane proteins. Taken together, we identified FYVE4 as a novel plant endosomal regulator, which functions in ESCRTing pathway to regulate MVE biogenesis.
Collapse
Affiliation(s)
- Chuanliang Liu
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yonglun Zeng
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Hongbo Li
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Chao Yang
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Wenjin Shen
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Min Xu
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Zhidan Xiao
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Tongsheng Chen
- MOE Key Laboratory & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Baiying Li
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wenhan Cao
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Liwen Jiang
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Marisa S Otegui
- Department of Botany, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Caiji Gao
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
44
|
Banjade S, Shah YH, Tang S, Emr SD. Design principles of the ESCRT-III Vps24-Vps2 module. eLife 2021; 10:67709. [PMID: 34028356 PMCID: PMC8143795 DOI: 10.7554/elife.67709] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022] Open
Abstract
ESCRT-III polymerization is required for all endosomal sorting complex required for transport (ESCRT)-dependent events in the cell. However, the relative contributions of the eight ESCRT-III subunits differ between each process. The minimal features of ESCRT-III proteins necessary for function and the role for the multiple ESCRT-III subunits remain unclear. To identify essential features of ESCRT-III subunits, we previously studied the polymerization mechanisms of two ESCRT-III subunits Snf7 and Vps24, identifying the association of the helix-4 region of Snf7 with the helix-1 region of Vps24 (Banjade et al., 2019a). Here, we find that mutations in the helix-1 region of another ESCRT-III subunit Vps2 can functionally replace Vps24 in Saccharomyces cerevisiae. Engineering and genetic selections revealed the required features of both subunits. Our data allow us to propose three minimal features required for ESCRT-III function – spiral formation, lateral association of the spirals through heteropolymerization, and binding to the AAA + ATPase Vps4 for dynamic remodeling.
Collapse
Affiliation(s)
- Sudeep Banjade
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Yousuf H Shah
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Shaogeng Tang
- Department of Biochemistry, Stanford University, Stanford, United States
| | - Scott D Emr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
45
|
Pfitzner AK, Moser von Filseck J, Roux A. Principles of membrane remodeling by dynamic ESCRT-III polymers. Trends Cell Biol 2021; 31:856-868. [PMID: 33980463 DOI: 10.1016/j.tcb.2021.04.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Endosomal protein complex required for transport-III (ESCRT-III) polymers are involved in many crucial cellular functions, from cell division to endosome-lysosome dynamics. As a eukaryotic membrane remodeling machinery, ESCRT-III is unique in its ability to catalyze fission of membrane necks from their luminal side and to participate in membrane remodeling processes of essentially all cellular organelles. Found in Archaea, it is also the most evolutionary ancient membrane remodeling machinery. The simple protein structure shared by all of its subunits assembles into a large variety of filament shapes, limiting our understanding of how these filaments achieve membrane remodeling. Here, we review recent findings that discovered unpredicted properties of ESCRT-III polymers, which enable us to define general principles of the mechanism by which ESCRT-III filaments remodel membranes.
Collapse
Affiliation(s)
| | | | - Aurélien Roux
- Biochemistry Department, University of Geneva, CH-1211 Geneva, Switzerland; Swiss National Centre for Competence in Research Programme Chemical Biology, CH-1211 Geneva, Switzerland.
| |
Collapse
|
46
|
Alqabandi M, de Franceschi N, Maity S, Miguet N, Bally M, Roos WH, Weissenhorn W, Bassereau P, Mangenot S. The ESCRT-III isoforms CHMP2A and CHMP2B display different effects on membranes upon polymerization. BMC Biol 2021; 19:66. [PMID: 33832485 PMCID: PMC8033747 DOI: 10.1186/s12915-021-00983-9] [Citation(s) in RCA: 20] [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: 01/23/2021] [Accepted: 02/16/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND ESCRT-III proteins are involved in many membrane remodeling processes including multivesicular body biogenesis as first discovered in yeast. In humans, ESCRT-III CHMP2 exists as two isoforms, CHMP2A and CHMP2B, but their physical characteristics have not been compared yet. RESULTS Here, we use a combination of techniques on biomimetic systems and purified proteins to study their affinity and effects on membranes. We establish that CHMP2B binding is enhanced in the presence of PI(4,5)P2 lipids. In contrast, CHMP2A does not display lipid specificity and requires CHMP3 for binding significantly to membranes. On the micrometer scale and at moderate bulk concentrations, CHMP2B forms a reticular structure on membranes whereas CHMP2A (+CHMP3) binds homogeneously. Thus, CHMP2A and CHMP2B unexpectedly induce different mechanical effects to membranes: CHMP2B strongly rigidifies them while CHMP2A (+CHMP3) has no significant effect. CONCLUSIONS We therefore conclude that CHMP2B and CHMP2A exhibit different mechanical properties and might thus contribute differently to the diverse ESCRT-III-catalyzed membrane remodeling processes.
Collapse
Affiliation(s)
- Maryam Alqabandi
- Laboratoire Physico Chimie Curie, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, 75005, Paris, France
| | - Nicola de Franceschi
- Laboratoire Physico Chimie Curie, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, 75005, Paris, France
| | - Sourav Maity
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Nolwenn Miguet
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38000, Grenoble, France
| | - Marta Bally
- Umeå University, Department of Clinical Microbiology & Wallenberg Centre for Molecular Medicine, 90185, Umeå, Sweden
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands
| | - Winfried Weissenhorn
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38000, Grenoble, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, 75005, Paris, France
| | - Stéphanie Mangenot
- Laboratoire Physico Chimie Curie, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, 75005, Paris, France.
| |
Collapse
|
47
|
Lu MS, Drubin DG. Cdc42 GTPase regulates ESCRTs in nuclear envelope sealing and ER remodeling. J Cell Biol 2021; 219:151867. [PMID: 32556066 PMCID: PMC7401818 DOI: 10.1083/jcb.201910119] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/11/2020] [Accepted: 05/01/2020] [Indexed: 12/21/2022] Open
Abstract
Small GTPases of the Rho family are binary molecular switches that regulate a variety of processes including cell migration and oriented cell divisions. Known Cdc42 effectors include proteins involved in cytoskeletal remodeling and kinase-dependent transcription induction, but none are involved in the maintenance of nuclear envelope integrity or ER morphology. Maintenance of nuclear envelope integrity requires the EndoSomal Complexes Required for Transport (ESCRT) proteins, but how they are regulated in this process remains unknown. Here, we show by live-cell imaging a novel Cdc42 localization with ESCRT proteins at sites of nuclear envelope and ER fission and, by genetic analysis of cdc42 mutant yeast, uncover a unique Cdc42 function in regulation of ESCRT proteins at the nuclear envelope and sites of ER tubule fission. Our findings implicate Cdc42 in nuclear envelope sealing and ER remodeling, where it regulates ESCRT disassembly to maintain nuclear envelope integrity and proper ER architecture.
Collapse
Affiliation(s)
- Michelle Seiko Lu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
48
|
Why Cells and Viruses Cannot Survive without an ESCRT. Cells 2021; 10:cells10030483. [PMID: 33668191 PMCID: PMC7995964 DOI: 10.3390/cells10030483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 12/15/2022] Open
Abstract
Intracellular organelles enwrapped in membranes along with a complex network of vesicles trafficking in, out and inside the cellular environment are one of the main features of eukaryotic cells. Given their central role in cell life, compartmentalization and mechanisms allowing their maintenance despite continuous crosstalk among different organelles have been deeply investigated over the past years. Here, we review the multiple functions exerted by the endosomal sorting complex required for transport (ESCRT) machinery in driving membrane remodeling and fission, as well as in repairing physiological and pathological membrane damages. In this way, ESCRT machinery enables different fundamental cellular processes, such as cell cytokinesis, biogenesis of organelles and vesicles, maintenance of nuclear–cytoplasmic compartmentalization, endolysosomal activity. Furthermore, we discuss some examples of how viruses, as obligate intracellular parasites, have evolved to hijack the ESCRT machinery or part of it to execute/optimize their replication cycle/infection. A special emphasis is given to the herpes simplex virus type 1 (HSV-1) interaction with the ESCRT proteins, considering the peculiarities of this interplay and the need for HSV-1 to cross both the nuclear-cytoplasmic and the cytoplasmic-extracellular environment compartmentalization to egress from infected cells.
Collapse
|
49
|
Meusser B, Purfuerst B, Luft FC. HIV-1 Gag release from yeast reveals ESCRT interaction with the Gag N-terminal protein region. J Biol Chem 2020; 295:17950-17972. [PMID: 32994219 PMCID: PMC7939435 DOI: 10.1074/jbc.ra120.014710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/25/2020] [Indexed: 11/30/2022] Open
Abstract
The HIV-1 protein Gag assembles at the plasma membrane and drives virion budding, assisted by the cellular endosomal complex required for transport (ESCRT) proteins. Two ESCRT proteins, TSG101 and ALIX, bind to the Gag C-terminal p6 peptide. TSG101 binding is important for efficient HIV-1 release, but how ESCRTs contribute to the budding process and how their activity is coordinated with Gag assembly is poorly understood. Yeast, allowing genetic manipulation that is not easily available in human cells, has been used to characterize the cellular ESCRT function. Previous work reported Gag budding from yeast spheroplasts, but Gag release was ESCRT-independent. We developed a yeast model for ESCRT-dependent Gag release. We combined yeast genetics and Gag mutational analysis with Gag-ESCRT binding studies and the characterization of Gag-plasma membrane binding and Gag release. With our system, we identified a previously unknown interaction between ESCRT proteins and the Gag N-terminal protein region. Mutations in the Gag-plasma membrane-binding matrix domain that reduced Gag-ESCRT binding increased Gag-plasma membrane binding and Gag release. ESCRT knockout mutants showed that the release enhancement was an ESCRT-dependent effect. Similarly, matrix mutation enhanced Gag release from human HEK293 cells. Release enhancement partly depended on ALIX binding to p6, although binding site mutation did not impair WT Gag release. Accordingly, the relative affinity for matrix compared with p6 in GST-pulldown experiments was higher for ALIX than for TSG101. We suggest that a transient matrix-ESCRT interaction is replaced when Gag binds to the plasma membrane. This step may activate ESCRT proteins and thereby coordinate ESCRT function with virion assembly.
Collapse
Affiliation(s)
- Birgit Meusser
- Charité Medical Faculty, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Friedrich C Luft
- Charité Medical Faculty, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Experimental and Clinical Research Center, Berlin, Germany.
| |
Collapse
|
50
|
Baeumers M, Ruhnau K, Breuer T, Pannen H, Goerlich B, Kniebel A, Haensch S, Weidtkamp-Peters S, Schmitt L, Klein T. Lethal (2) giant discs (Lgd)/CC2D1 is required for the full activity of the ESCRT machinery. BMC Biol 2020; 18:200. [PMID: 33349255 PMCID: PMC7754597 DOI: 10.1186/s12915-020-00933-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/24/2020] [Indexed: 01/10/2023] Open
Abstract
Background A major task of the endosomal sorting complex required for transport (ESCRT) machinery is the pinching off of cargo-loaded intraluminal vesicles (ILVs) into the lumen of maturing endosomes (MEs), which is essential for the complete degradation of transmembrane proteins in the lysosome. The ESCRT machinery is also required for the termination of signalling through activated signalling receptors, as it separates their intracellular domains from the cytosol. At the heart of the machinery lies the ESCRT-III complex, which is required for an increasing number of processes where membrane regions are abscised away from the cytosol. The core of ESCRT-III, comprising four members of the CHMP protein family, organises the assembly of a homopolymer of CHMP4, Shrub in Drosophila, that is essential for abscission. We and others identified the tumour-suppressor lethal (2) giant discs (Lgd)/CC2D1 as a physical interactor of Shrub/CHMP4 in Drosophila and mammals, respectively. Results Here, we show that the loss of function of lgd constitutes a state of reduced activity of Shrub/CHMP4/ESCRT-III. This hypomorphic shrub mutant situation causes a slight decrease in the rate of ILV formation that appears to result in incomplete incorporation of Notch into ILVs. We found that the forced incorporation in ILVs of lgd mutant MEs suppresses the uncontrolled and ligand-independent activation of Notch. Moreover, the analysis of Su(dx) lgd double mutants clarifies their relationship and suggests that they are not operating in a linear pathway. We could show that, despite prolonged lifetime, the MEs of lgd mutants have a similar ILV density as wild-type but less than rab7 mutant MEs, suggesting the rate in lgd mutants is slightly reduced. The analysis of the MEs of wild-type and mutant cells in the electron microscope revealed that the ESCRT-containing electron-dense microdomains of ILV formation at the limiting membrane are elongated, indicating a change in ESCRT activity. Since lgd mutants can be rescued to normal adult flies if extra copies of shrub (or its mammalian ortholog CHMP4B) are added into the genome, we conclude that the net activity of Shrub is reduced upon loss of lgd function. Finally, we show that, in solution, CHMP4B/Shrub exists in two conformations. LGD1/Lgd binding does not affect the conformational state of Shrub, suggesting that Lgd is not a chaperone for Shrub/CHMP4B. Conclusion Our results suggest that Lgd is required for the full activity of Shrub/ESCRT-III. In its absence, the activity of the ESCRT machinery is reduced. This reduction causes the escape of a fraction of cargo, among it Notch, from incorporation into ILVs, which in turn leads to an activation of this fraction of Notch after fusion of the ME with the lysosome. Our results highlight the importance of the incorporation of Notch into ILV not only to assure complete degradation, but also to avoid uncontrolled activation of the pathway.
Collapse
Affiliation(s)
- Miriam Baeumers
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Kristina Ruhnau
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Thomas Breuer
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Hendrik Pannen
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Bastian Goerlich
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Anna Kniebel
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Sebastian Haensch
- Center of Advanced Imaging (CAi), Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Stefanie Weidtkamp-Peters
- Center of Advanced Imaging (CAi), Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Lutz Schmitt
- Institute of Biochemistry I, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany.
| |
Collapse
|