1
|
Breuer T, Tibbe C, Troost T, Klein T. Structural Analysis of the ESCRT-III Regulator Lethal(2) Giant Discs/Coiled-Coil and C2 Domain-Containing Protein 1 (Lgd/CC2D1). Cells 2024; 13:1174. [PMID: 39056756 PMCID: PMC11275157 DOI: 10.3390/cells13141174] [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: 06/03/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Members of the LGD/CC2D1 protein family contain repeats of the family-defining DM14 domains. Via this domain, they interact with members of the CHMP family, which are essential for the ESCRT machinery-mediated formation of intraluminal vesicles during endosome maturation. Here, we investigate the requirement of the DM14 domains for the function of Lgd in detail. We found that although both odd-numbered DM14s can act in a functionally redundant manner, the redundancy is not complete and both contribute to the full function of Lgd. Our analysis indicates that some of the AAs that form the KARRxxR motif of the onDM14s are not exchangeable by similarly charged AAs without loss of function, indicating that they not only provide charge, but also fulfil structural roles. Furthermore, we show that the region of Lgd between DM14-4 and the C2 domain as well as its C-terminal region to the C2 domain are important for protein stability/function. Moreover, we analysed the importance of AAs that are conserved in all DM14 domains. Finally, our analysis of the C. elegans ortholog of Lgd revealed that it has only one DM14 domain that is functionally equivalent to the onDM14s. Altogether, the results further the understanding of how Lgd family members regulate the ESCRT machinery.
Collapse
Affiliation(s)
| | | | | | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany; (T.B.); (C.T.); (T.T.)
| |
Collapse
|
2
|
Prelli Bozzo C, Laliberté A, De Luna A, Pastorio C, Regensburger K, Krebs S, Graf A, Blum H, Volcic M, Sparrer KMJ, Kirchhoff F. Replication competent HIV-guided CRISPR screen identifies antiviral factors including targets of the accessory protein Nef. Nat Commun 2024; 15:3813. [PMID: 38714682 PMCID: PMC11076291 DOI: 10.1038/s41467-024-48228-x] [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: 12/06/2023] [Accepted: 04/24/2024] [Indexed: 05/10/2024] Open
Abstract
Innate antiviral factors are essential for effective defense against viral pathogens. However, the identity of major restriction mechanisms remains elusive. Current approaches to discover antiviral factors usually focus on the initial steps of viral replication and are limited to a single round of infection. Here, we engineered libraries of >1500 replication-competent HIV-1 constructs each expressing a single gRNAs to target >500 cellular genes for virus-driven discovery of antiviral factors. Passaging in CD4+ T cells robustly enriched HIV-1 encoding sgRNAs against GRN, CIITA, EHMT2, CEACAM3, CC2D1B and RHOA by >50-fold. Using an HIV-1 library lacking the accessory nef gene, we identified IFI16 as a Nef target. Functional analyses in cell lines and primary CD4+ T cells support that the HIV-driven CRISPR screen identified restriction factors targeting virus entry, transcription, release and infectivity. Our HIV-guided CRISPR technique enables sensitive discovery of physiologically relevant cellular defense factors throughout the entire viral replication cycle.
Collapse
Affiliation(s)
| | - Alexandre Laliberté
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Aurora De Luna
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Chiara Pastorio
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Kerstin Regensburger
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis Gene Center, LMU Munich, 81377, Munich, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis Gene Center, LMU Munich, 81377, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis Gene Center, LMU Munich, 81377, Munich, Germany
| | - Meta Volcic
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
3
|
Xie T, Feng M, Zhang X, Li X, Mo G, Shi M, Zhang X. Chicken CH25H inhibits ALV-J replication by promoting cellular autophagy. Front Immunol 2023; 14:1093289. [PMID: 36875122 PMCID: PMC9975585 DOI: 10.3389/fimmu.2023.1093289] [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: 11/08/2022] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Autophagy plays an important role in host antiviral defense. The avian leukosis virus subgroup J (ALV-J) has been shown to inhibit autophagy while promoting viral replication. The underlying autophagic mechanisms, however, are unknown. Cholesterol 25-hydroxylase (CH25H) is a conserved interferon-stimulated gene, which converts cholesterol to a soluble antiviral factor, 25-hydroxycholesterol (25HC). In this study, we further investigated the autophagic mechanism of CH25H resistance to ALV-J in chicken embryonic fibroblast cell lines (DF1). Our results found that overexpression of CH25H and treatment with 25HC promoted the autophagic markers microtubule-associated protein 1 light chain 3 II (LC3II) and autophagy-related gene 5(ATG5), while decreased autophagy substrate p62/SQSTM1 (p62) expression in ALV-J infection DF-1 cells. Induction of cellular autophagy also reduces the levels of ALV-J gp85 and p27. ALV-J infection, on the other hand, suppresses autophagic marker protein LC3II expression. These findings suggest that CH25H-induced autophagy is a host defense mechanism that aids in ALV-J replication inhibition. In particular, CH25H interacts with CHMP4B and inhibits ALV-J infection in DF-1 cells by promoting autophagy, revealing a novel mechanism by which CH25H inhibits ALV-J infection. Although the underlying mechanisms are not completely understood, CH25H and 25HC are the first to show inhibiting ALV-J infection via autophagy.
Collapse
Affiliation(s)
- Tingting Xie
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xi Zhang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiaoqi Li
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| | - Guodong Mo
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Xiquan Zhang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Clarke AL, Lettman MM, Audhya A. Lgd regulates ESCRT-III complex accumulation at multivesicular endosomes to control intralumenal vesicle formation. Mol Biol Cell 2022; 33:ar144. [PMID: 36287829 PMCID: PMC9727795 DOI: 10.1091/mbc.e22-08-0342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 02/04/2023] Open
Abstract
Membrane remodeling mediated by heteropolymeric filaments composed of ESCRT-III subunits is an essential process that occurs at a variety of organelles to maintain cellular homeostasis. Members of the evolutionarily conserved Lgd/CC2D1 protein family have been suggested to regulate ESCRT-III polymer assembly, although their specific roles, particularly in vivo, remain unclear. Using the Caenorhabditis elegans early embryo as a model system, we show that Lgd/CC2D1 localizes to endosomal membranes, and its loss impairs endolysosomal cargo sorting and degradation. At the ultrastructural level, the absence of Lgd/CC2D1 results in the accumulation of enlarged endosomal compartments that contain a reduced number of intralumenal vesicles (ILVs). However, unlike aberrant endosome morphology caused by depletion of other ESCRT components, ILV size is only modestly altered in embryos lacking Lgd/CC2D1. Instead, loss of Lgd/CC2D1 impairs normal accumulation of ESCRT-III on endosomal membranes, likely slowing the kinetics of ILV formation. Together, our findings suggest a role for Lgd/CC2D1 in the recruitment and/or stable assembly of ESCRT-III subunits on endosomal membranes to facilitate efficient ILV biogenesis.
Collapse
Affiliation(s)
- Aryel L. Clarke
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Molly M. Lettman
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| |
Collapse
|
5
|
Baeumers M, Schulz K, Klein T. Using Drosophila melanogaster to Analyse the Human Paralogs of the ESCRT-III Core Component Shrub/CHMP4/Snf7 and Its Interactions with Members of the LGD/CC2D1 Family. Int J Mol Sci 2022; 23:7507. [PMID: 35886850 PMCID: PMC9320689 DOI: 10.3390/ijms23147507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
The evolutionary conserved ESCRT-III complex is a device for membrane remodelling in various cellular processes, such as the formation of intraluminal vesicles (ILVs), cytokinesis, and membrane repair. The common theme of all these processes is the abscission of membrane away from the cytosol. At its heart in Drosophila is Shrub, CHMP4 in humans, which dynamically polymerises into filaments through electrostatic interactions among the protomers. For the full activity, Shrub/CHMP4 requires physical interaction with members of the Lgd protein family. This interaction is mediated by the odd-numbered DM14 domains of Lgd, which bind to the negative interaction surface of Shrub. While only one Lgd and one Shrub exist in the genome of Drosophila, mammals have two Lgd orthologs, LGD1/CC2D1B and LGD2/CC2D1A, as well as three CHMP4s in their genomes, CHMP4A, CHMP4B, and CHMP4C. The rationale for the diversification of the ESCRT components is not understood. We here use Drosophila as a model system to analyse the activity of the human orthologs of Shrub and Lgd at an organismal level. This enabled us to use the plethora of available techniques available for Drosophila. We present evidence that CHMP4B is the true ortholog of Shrub, while CHMP4A and CHMP4C have diverging activities. Nevertheless, CHMP4A and CHMP4C can enhance the activity of CHMP4B, raising the possibility that they can form heteropolymers in vivo. Our structure-function analysis of the LGD1 and LGD2 indicates that the C2 domain of the LGD proteins has a specific function beyond protein stability and subcellular localisation. Moreover, our data specify that CHMP4B interacts more efficiently with LGD1 than with LGD2.
Collapse
Affiliation(s)
- Miriam Baeumers
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany;
| | - Katharina Schulz
- Institut für Angewandte Bewegungswissenschaften, Professur für Sportmedizin/-Biologie, Technische Universität Chemnitz, Thüringer Weg 11, 09126 Chemnitz, Germany;
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany;
| |
Collapse
|
6
|
Proximity-Labeling Reveals Novel Host and Parasite Proteins at the Toxoplasma Parasitophorous Vacuole Membrane. mBio 2021; 12:e0026021. [PMID: 34749525 PMCID: PMC8576527 DOI: 10.1128/mbio.00260-21] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii is a ubiquitous, intracellular parasite that envelops its parasitophorous vacuole with a protein-laden membrane (PVM). The PVM is critical for interactions with the infected host cell, such as nutrient transport and immune defense. Only a few parasite and host proteins have so far been identified on the host-cytosolic side of the Toxoplasma PVM. We report here the use of human foreskin fibroblasts expressing the proximity-labeling enzyme miniTurbo, fused to a domain that targets it to this face of the PVM, in combination with quantitative proteomics to specifically identify proteins present at this interface. Out of numerous human and parasite proteins with candidate PVM localization, we validate three parasite proteins (TGGT1_269950 [GRA61], TGGT1_215360 [GRA62], and TGGT1_217530 [GRA63]) and four new host proteins (PDCD6IP/ALIX, PDCD6, CC2D1A, and MOSPD2) as localized to the PVM in infected human cells through immunofluorescence microscopy. These results significantly expand our knowledge of proteins present at the Toxoplasma PVM and, given that three of the validated host proteins are components of the ESCRT (endosomal sorting complexes required for transport) machinery, they further suggest that novel biology is operating at this crucial host-pathogen interface.
Collapse
|
7
|
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
|
8
|
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
|
9
|
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: 6] [Impact Index Per Article: 1.2] [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
|
10
|
Ma ACH, Mak CCY, Yeung KS, Pei SLC, Ying D, Yu MHC, Hasan KMM, Chen X, Chow PC, Cheung YF, Chung BHY. Monoallelic Mutations in CC2D1A Suggest a Novel Role in Human Heterotaxy and Ciliary Dysfunction. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e003000. [PMID: 33196317 PMCID: PMC7748040 DOI: 10.1161/circgen.120.003000] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Human heterotaxy is a group of congenital disorders characterized by misplacement of one or more organs according to the left-right axis. The genetic causes of human heterotaxy are highly heterogeneous. METHODS We performed exome sequencing in a cohort of 26 probands with heterotaxy followed by gene burden analysis for the enrichment of novel rare damaging mutations. Transcription activator-like effector nuclease was used to generate somatic loss-of-function mutants in a zebrafish model. Ciliary defects were examined by whole-mount immunostaining of acetylated α-tubulin. RESULTS We identified a significant enrichment of novel rare damaging mutations in the CC2D1A gene. Seven occurrences of CC2D1A mutations were found to affect 4 highly conserved amino acid residues of the protein. Functional analyses in the transcription activator-like effector nuclease-mediated zebrafish knockout models were performed, and heterotaxy phenotypes of the cardiovascular and gastrointestinal systems in both somatic and germline mutants were observed. Defective cilia were demonstrated by whole-mount immunostaining of acetylated α-tubulin. These abnormalities were rescued by wild-type cc2d1a mRNA but not cc2d1a mutant mRNA, strongly suggesting a loss-of-function mechanism. On the other hand, overexpression of cc2d1a orthologous mutations cc2d1a P559L and cc2d1a G808V (orthologous to human CC2D1A P532L and CC2D1A G781V) did not affect embryonic development. CONCLUSIONS Using a zebrafish model, we were able to establish a novel association of CC2D1A with heterotaxy and ciliary dysfunction in the F2 generation via a loss-of-function mechanism. Future mechanistic studies are needed for a better understanding of the role of CC2D1A in left-right patterning and ciliary dysfunction.
Collapse
Affiliation(s)
- Alvin Chun Hang Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrate Region, China (A.C.H., K.M.M.H.)
| | - Christopher Chun Yu Mak
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Kit San Yeung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Steven Lim Cho Pei
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Dingge Ying
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Mullin Ho Chung Yu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Kazi Md Mahmudul Hasan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrate Region, China (A.C.H., K.M.M.H.)
| | - Xiangke Chen
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China (X.C.)
| | - Pak Cheong Chow
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Yiu Fai Cheung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| | - Brian Hon Yin Chung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Kong Kong, Hong Kong Special Administrate Region, China (C.C.Y.M., K.S.Y., S.L.C.P., D.Y., M.H.C.Y., P.C.C., Y.F.C., B.H.Y.C.)
| |
Collapse
|
11
|
Butt BG, Owen DJ, Jeffries CM, Ivanova L, Hill CH, Houghton JW, Ahmed MF, Antrobus R, Svergun DI, Welch JJ, Crump CM, Graham SC. Insights into herpesvirus assembly from the structure of the pUL7:pUL51 complex. eLife 2020; 9:e53789. [PMID: 32391791 PMCID: PMC7289601 DOI: 10.7554/elife.53789] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/07/2020] [Indexed: 12/19/2022] Open
Abstract
Herpesviruses acquire their membrane envelopes in the cytoplasm of infected cells via a molecular mechanism that remains unclear. Herpes simplex virus (HSV)-1 proteins pUL7 and pUL51 form a complex required for efficient virus envelopment. We show that interaction between homologues of pUL7 and pUL51 is conserved across human herpesviruses, as is their association with trans-Golgi membranes. We characterized the HSV-1 pUL7:pUL51 complex by solution scattering and chemical crosslinking, revealing a 1:2 complex that can form higher-order oligomers in solution, and we solved the crystal structure of the core pUL7:pUL51 heterodimer. While pUL7 adopts a previously-unseen compact fold, the helix-turn-helix conformation of pUL51 resembles the cellular endosomal complex required for transport (ESCRT)-III component CHMP4B and pUL51 forms ESCRT-III-like filaments, suggesting a direct role for pUL51 in promoting membrane scission during virus assembly. Our results provide a structural framework for understanding the role of the conserved pUL7:pUL51 complex in herpesvirus assembly.
Collapse
Affiliation(s)
- Benjamin G Butt
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Danielle J Owen
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Cy M Jeffries
- European Molecular Biology Laboratory (EMBL) Hamburg SiteHamburgGermany
| | - Lyudmila Ivanova
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Chris H Hill
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Jack W Houghton
- Cambridge Institute for Medical Research, University of CambridgeCambridgeUnited Kingdom
| | - Md Firoz Ahmed
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of CambridgeCambridgeUnited Kingdom
| | - Dmitri I Svergun
- European Molecular Biology Laboratory (EMBL) Hamburg SiteHamburgGermany
| | - John J Welch
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
| | - Colin M Crump
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| | - Stephen C Graham
- Department of Pathology, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
12
|
Abstract
Cellular membranes can form two principally different involutions, which either exclude or contain cytosol. The 'classical' budding reactions, such as those occurring during endocytosis or formation of exocytic vesicles, involve proteins that assemble on the cytosol-excluding face of the bud neck. Inverse membrane involution occurs in a wide range of cellular processes, supporting cytokinesis, endosome maturation, autophagy, membrane repair and many other processes. Such inverse membrane remodelling is mediated by a heteromultimeric protein machinery known as endosomal sorting complex required for transport (ESCRT). ESCRT proteins assemble on the cytosolic (or nucleoplasmic) face of the neck of the forming involution and cooperate with the ATPase VPS4 to drive membrane scission or sealing. Here, we review similarities and differences of various ESCRT-dependent processes, with special emphasis on mechanisms of ESCRT recruitment.
Collapse
|
13
|
Liao YT, Lin SS, Lin SJ, Sun WT, Shen BN, Cheng HP, Lin CP, Ko TP, Chen YF, Wang HC. Structural insights into the interaction between phytoplasmal effector causing phyllody 1 and MADS transcription factors. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2019; 100:706-719. [PMID: 31323156 DOI: 10.1111/tpj.14463] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/24/2019] [Accepted: 07/02/2019] [Indexed: 05/21/2023]
Abstract
Phytoplasmas are bacterial plant pathogens which can induce severe symptoms including dwarfism, phyllody and virescence in an infected plant. Because phytoplasmas infect many important crops such as peanut and papaya they have caused serious agricultural losses. The phytoplasmal effector causing phyllody 1 (PHYL1) is an important phytoplasmal pathogenic factor which affects the biological function of MADS transcription factors by interacting with their K (keratin-like) domain, thus resulting in abnormal plant developments such as phyllody. Until now, lack of information on the structure of PHYL1 has prevented a detailed understanding of the binding mechanism between PHYL1 and the MADS transcription factors. Here, we present the crystal structure of PHYL1 from peanut witches'-broom phytoplasma (PHYL1PnWB ). This protein was found to fold into a unique α-helical hairpin with exposed hydrophobic residues on its surface that may play an important role in its biological function. Using proteomics approaches, we propose a binding mode of PHYL1PnWB with the K domain of the MADS transcription factor SEPALLATA3 (SEP3_K) and identify the residues of PHYL1PnWB that are important for this interaction. Furthermore, using surface plasmon resonance we measure the binding strength of PHYL1PnWB proteins to SEP3_K. Lastly, based on confocal images, we found that α-helix 2 of PHYL1PnWB plays an important role in PHYL1-mediated degradation of SEP3. Taken together, these results provide a structural understanding of the specific binding mechanism between PHYL1PnWB and SEP3_K.
Collapse
Affiliation(s)
- Yi-Ting Liao
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Shih-Shun Lin
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
- Center of Biotechnology, National Taiwan University, Taipei 10617, Taiwan
| | - Shin-Jen Lin
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan
| | - Wan-Ting Sun
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Department of Plant Pathology and Microbiology, College of Bioresources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
| | - Bing-Nan Shen
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Department of Plant Pathology and Microbiology, College of Bioresources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
| | - Han-Pin Cheng
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Department of Plant Pathology and Microbiology, College of Bioresources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
| | - Chan-Pin Lin
- Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Department of Plant Pathology and Microbiology, College of Bioresources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
| | - Tzu-Ping Ko
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Fan Chen
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Hao-Ching Wang
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
14
|
Gatta AT, Carlton JG. The ESCRT-machinery: closing holes and expanding roles. Curr Opin Cell Biol 2019; 59:121-132. [DOI: 10.1016/j.ceb.2019.04.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 01/08/2023]
|
15
|
Vietri M, Stenmark H. Orchestrating Nuclear Envelope Sealing during Mitosis. Dev Cell 2019; 47:541-542. [PMID: 30513298 DOI: 10.1016/j.devcel.2018.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sealing of the nascent nuclear envelope during mitotic exit is mediated by the polymeric ESCRT-III protein machinery. In this issue of Developmental Cell, Ventimiglia et al. (2018) identify the ESCRT-III-associated protein CC2D1B as a lipid-binding coordinator of late-mitotic ESCRT-III assembly and disassembly of the mitotic spindle.
Collapse
Affiliation(s)
- Marina Vietri
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, N-0379 Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway
| | - Harald Stenmark
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, N-0379 Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway.
| |
Collapse
|
16
|
The role of VPS4 in ESCRT-III polymer remodeling. Biochem Soc Trans 2019; 47:441-448. [DOI: 10.1042/bst20180026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 01/04/2023]
Abstract
Abstract
The endosomal sorting complex required for transport-III (ESCRT-III) and VPS4 catalyze a variety of membrane-remodeling processes in eukaryotes and archaea. Common to these processes is the dynamic recruitment of ESCRT-III proteins from the cytosol to the inner face of a membrane neck structure, their activation and filament formation inside or at the membrane neck and the subsequent or concomitant recruitment of the AAA-type ATPase VPS4. The dynamic assembly of ESCRT-III filaments and VPS4 on cellular membranes induces constriction of membrane necks with large diameters such as the cytokinetic midbody and necks with small diameters such as those of intraluminal vesicles or enveloped viruses. The two processes seem to use different sets of ESCRT-III filaments. Constriction is then thought to set the stage for membrane fission. Here, we review recent progress in understanding the structural transitions of ESCRT-III proteins required for filament formation, the functional role of VPS4 in dynamic ESCRT-III assembly and its active role in filament constriction. The recent data will be discussed in the context of different mechanistic models for inside-out membrane fission.
Collapse
|
17
|
Ventimiglia LN, Cuesta-Geijo MA, Martinelli N, Caballe A, Macheboeuf P, Miguet N, Parnham IM, Olmos Y, Carlton JG, Weissenhorn W, Martin-Serrano J. CC2D1B Coordinates ESCRT-III Activity during the Mitotic Reformation of the Nuclear Envelope. Dev Cell 2018; 47:547-563.e6. [PMID: 30513301 PMCID: PMC6286407 DOI: 10.1016/j.devcel.2018.11.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 08/13/2018] [Accepted: 11/02/2018] [Indexed: 11/29/2022]
Abstract
The coordinated reformation of the nuclear envelope (NE) after mitosis re-establishes the structural integrity and the functionality of the nuclear compartment. The endosomal sorting complex required for transport (ESCRT) machinery, a membrane remodeling pathway that is highly conserved in eukaryotes, has been recently involved in NE resealing by mediating the annular fusion of the nuclear membrane (NM). We show here that CC2D1B, a regulator of ESCRT polymerization, is required to re-establish the nuclear compartmentalization by coordinating endoplasmic reticulum (ER) membrane deposition around chromatin disks with ESCRT-III recruitment to the reforming NE. Accordingly, CC2D1B determines the spatiotemporal distribution of the CHMP7-ESCRT-III axis during NE reformation. Crucially, in CC2D1B-depleted cells, ESCRT activity is uncoupled from Spastin-mediated severing of spindle microtubules, resulting in persisting microtubules that compromise nuclear morphology. Therefore, we reveal CC2D1B as an essential regulatory factor that licenses the formation of ESCRT-III polymers to ensure the orderly reformation of the NE.
Collapse
Affiliation(s)
- Leandro N Ventimiglia
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London SE1 9RT, UK
| | - Miguel Angel Cuesta-Geijo
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London SE1 9RT, UK
| | - Nicolas Martinelli
- CNRS, University Grenoble Alpes, CEA, Institut de Biologie Structurale (IBS), 38000 Grenoble, France
| | - Anna Caballe
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London SE1 9RT, UK
| | - Pauline Macheboeuf
- CNRS, University Grenoble Alpes, CEA, Institut de Biologie Structurale (IBS), 38000 Grenoble, France
| | - Nolwenn Miguet
- CNRS, University Grenoble Alpes, CEA, Institut de Biologie Structurale (IBS), 38000 Grenoble, France
| | - Ian M Parnham
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London SE1 9RT, UK
| | - Yolanda Olmos
- Division of Cancer Studies, King's College London, London SE1 1UL, UK
| | - Jeremy G Carlton
- Division of Cancer Studies, King's College London, London SE1 1UL, UK
| | - Winfried Weissenhorn
- CNRS, University Grenoble Alpes, CEA, Institut de Biologie Structurale (IBS), 38000 Grenoble, France
| | - Juan Martin-Serrano
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London SE1 9RT, UK.
| |
Collapse
|
18
|
McCullough J, Frost A, Sundquist WI. Structures, Functions, and Dynamics of ESCRT-III/Vps4 Membrane Remodeling and Fission Complexes. Annu Rev Cell Dev Biol 2018; 34:85-109. [PMID: 30095293 PMCID: PMC6241870 DOI: 10.1146/annurev-cellbio-100616-060600] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRT) pathway mediates cellular membrane remodeling and fission reactions. The pathway comprises five core complexes: ALIX, ESCRT-I, ESCRT-II, ESCRT-III, and Vps4. These soluble complexes are typically recruited to target membranes by site-specific adaptors that bind one or both of the early-acting ESCRT factors: ALIX and ESCRT-I/ESCRT-II. These factors, in turn, nucleate assembly of ESCRT-III subunits into membrane-bound filaments that recruit the AAA ATPase Vps4. Together, ESCRT-III filaments and Vps4 remodel and sever membranes. Here, we review recent advances in our understanding of the structures, activities, and mechanisms of the ESCRT-III and Vps4 machinery, including the first high-resolution structures of ESCRT-III filaments, the assembled Vps4 enzyme in complex with an ESCRT-III substrate, the discovery that ESCRT-III/Vps4 complexes can promote both inside-out and outside-in membrane fission reactions, and emerging mechanistic models for ESCRT-mediated membrane fission.
Collapse
Affiliation(s)
- John McCullough
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA;
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Wesley I Sundquist
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA;
| |
Collapse
|
19
|
McMillan BJ, Tibbe C, Drabek AA, Seegar TCM, Blacklow SC, Klein T. Structural Basis for Regulation of ESCRT-III Complexes by Lgd. Cell Rep 2018; 19:1750-1757. [PMID: 28564595 DOI: 10.1016/j.celrep.2017.05.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/04/2017] [Accepted: 05/07/2017] [Indexed: 11/17/2022] Open
Abstract
The ESCRT-III complex induces outward membrane budding and fission through homotypic polymerization of its core component Shrub/CHMP4B. Shrub activity is regulated by its direct interaction with a protein called Lgd in flies, or CC2D1A or B in humans. Here, we report the structural basis for this interaction and propose a mechanism for regulation of polymer assembly. The isolated third DM14 repeat of Lgd binds Shrub, and an Lgd fragment containing only this DM14 repeat and its C-terminal C2 domain is sufficient for in vivo function. The DM14 domain forms a helical hairpin with a conserved, positively charged tip, that, in the structure of a DM14 domain-Shrub complex, occupies a negatively charged surface of Shrub that is otherwise used for homopolymerization. Lgd mutations at this interface disrupt its function in flies, confirming functional importance. Together, these data argue that Lgd regulates ESCRT activity by controlling access to the Shrub self-assembly surface.
Collapse
Affiliation(s)
- Brian J McMillan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Christine Tibbe
- Institute of Genetics, Heinrich-Heine-University, Dusseldorf 40225, Germany
| | - Andrew A Drabek
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Tom C M Seegar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Dana Farber Cancer Institute, Department of Cancer Biology, Boston, MA 02215, USA.
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-University, Dusseldorf 40225, Germany.
| |
Collapse
|
20
|
Zamarbide M, Oaks AW, Pond HL, Adelman JS, Manzini MC. Loss of the Intellectual Disability and Autism Gene Cc2d1a and Its Homolog Cc2d1b Differentially Affect Spatial Memory, Anxiety, and Hyperactivity. Front Genet 2018; 9:65. [PMID: 29552027 PMCID: PMC5840150 DOI: 10.3389/fgene.2018.00065] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/15/2018] [Indexed: 11/13/2022] Open
Abstract
Hundreds of genes are mutated in non-syndromic intellectual disability (ID) and autism spectrum disorder (ASD), with each gene often involved in only a handful of cases. Such heterogeneity can be daunting, but rare recessive loss of function (LOF) mutations can be a good starting point to provide insight into the mechanisms of neurodevelopmental disease. Biallelic LOF mutations in the signaling scaffold CC2D1A cause a rare form of autosomal recessive ID, sometimes associated with ASD and seizures. In parallel, we recently reported that Cc2d1a-deficient mice present with cognitive and social deficits, hyperactivity and anxiety. In Drosophila, loss of the only ortholog of Cc2d1a, lgd, is embryonically lethal, while in vertebrates, Cc2d1a has a homolog Cc2d1b which appears to be compensating, indicating that Cc2d1a and Cc2d1b have a redundant function in humans and mice. Here, we generate an allelic series of Cc2d1a and Cc2d1b LOF to determine the relative role of these genes during behavioral development. We generated Cc2d1b knockout (KO), Cc2d1a/1b double heterozygous and double KO mice, then performed behavioral studies to analyze learning and memory, social interactions, anxiety, and hyperactivity. We found that Cc2d1a and Cc2d1b have partially overlapping roles. Overall, loss of Cc2d1b is less severe than loss of Cc2d1a, only leading to cognitive deficits, while Cc2d1a/1b double heterozygous animals are similar to Cc2d1a-deficient mice. These results will help us better understand the deficits in individuals with CC2D1A mutations, suggesting that recessive CC2D1B mutations and trans-heterozygous CC2D1A and CC2D1B mutations could also contribute to the genetics of ID.
Collapse
Affiliation(s)
- Marta Zamarbide
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Adam W. Oaks
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Heather L. Pond
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Julia S. Adelman
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - M. Chiara Manzini
- GW Institute for Neurosciences, Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
- Autism and Neurodevelopmental Disorders Institute, The George Washington University, Washington, DC, United States
| |
Collapse
|
21
|
Avalos-Padilla Y, Knorr RL, Javier-Reyna R, García-Rivera G, Lipowsky R, Dimova R, Orozco E. The Conserved ESCRT-III Machinery Participates in the Phagocytosis of Entamoeba histolytica. Front Cell Infect Microbiol 2018; 8:53. [PMID: 29546036 PMCID: PMC5838018 DOI: 10.3389/fcimb.2018.00053] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/12/2018] [Indexed: 01/22/2023] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) orchestrates cell membrane-remodeling mechanisms in eukaryotes, including endocytosis. However, ESCRT functions in phagocytosis (ingestion of ≥250 nm particles), has been poorly studied. In macrophages and amoebae, phagocytosis is required for cell nutrition and attack to other microorganisms and cells. In Entamoeba histolytica, the voracious protozoan responsible for human amoebiasis, phagocytosis is a land mark of virulence. Here, we have investigated the role of ESCRT-III in the phagocytosis of E. histolytica, using mutant trophozoites, recombinant proteins (rEhVps20, rEhVps32, rEhVps24, and rEhVps2) and giant unilamellar vesicles (GUVs). Confocal images displayed the four proteins located around the ingested erythrocytes, in erythrocytes-containing phagosomes and in multivesicular bodies. EhVps32 and EhVps2 proteins co-localized at the phagocytic cups. Protein association increased during phagocytosis. Immunoprecipitation and flow cytometry assays substantiated these associations. GUVs revealed that the protein assembly sequence is essential to form intraluminal vesicles (ILVs). First, the active rEhVps20 bound to membranes and recruited rEhVps32, promoting membrane invaginations. rEhVps24 allowed the detachment of nascent vesicles, forming ILVs; and rEhVps2 modulated their size. The knock down of Ehvps20 and Ehvps24genes diminished the rate of erythrophagocytosis demonstrating the importance of ESCRT-III in this event. In conclusion, we present here evidence of the ESCRT-III participation in phagocytosis and delimitate the putative function of proteins, according to the in vitro reconstruction of their assembling.
Collapse
Affiliation(s)
- Yunuen Avalos-Padilla
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.,Departamento de Infectómica y Patogénesis Molecular, CINVESTAV IPN, Mexico City, Mexico
| | - Roland L Knorr
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Rosario Javier-Reyna
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV IPN, Mexico City, Mexico
| | | | - Reinhard Lipowsky
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Rumiana Dimova
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV IPN, Mexico City, Mexico
| |
Collapse
|
22
|
Foster TL, Pickering S, Neil SJD. Inhibiting the Ins and Outs of HIV Replication: Cell-Intrinsic Antiretroviral Restrictions at the Plasma Membrane. Front Immunol 2018; 8:1853. [PMID: 29354117 PMCID: PMC5758531 DOI: 10.3389/fimmu.2017.01853] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023] Open
Abstract
Like all viruses, human immunodeficiency viruses (HIVs) and their primate lentivirus relatives must enter cells in order to replicate and, once produced, new virions need to exit to spread to new targets. These processes require the virus to cross the plasma membrane of the cell twice: once via fusion mediated by the envelope glycoprotein to deliver the viral core into the cytosol; and secondly by ESCRT-mediated scission of budding virions during release. This physical barrier thus presents a perfect location for host antiviral restrictions that target enveloped viruses in general. In this review we will examine the current understanding of innate host antiviral defences that inhibit these essential replicative steps of primate lentiviruses associated with the plasma membrane, the mechanism by which these viruses have adapted to evade such defences, and the role that this virus/host battleground plays in the transmission and pathogenesis of HIV/AIDS.
Collapse
Affiliation(s)
- Toshana L Foster
- Department of Infectious Disease, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Suzanne Pickering
- Department of Infectious Disease, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Stuart J D Neil
- Department of Infectious Disease, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
23
|
Alfred V, Vaccari T. Mechanisms of Non-canonical Signaling in Health and Disease: Diversity to Take Therapy up a Notch? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:187-204. [PMID: 30030827 DOI: 10.1007/978-3-319-89512-3_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Non-canonical Notch signaling encompasses a wide range of cellular processes, diverging considerably from the established paradigm. It can dispense of ligand, proteolytic or nuclear activity. Non-canonical Notch signaling events have been studied mostly in the fruit fly Drosophila melanogaster, the organism in which Notch was identified first and a powerful model for understanding signaling outcomes. However, non-canonical events are ill-defined and their involvement in human physiology is not clear, hampering our understanding of diseases arising from Notch signaling alterations. At a time in which therapies based on specific targeting of Notch signaling are still an unfulfilled promise, detailed understanding of non-canonical Notch events might be key to devising more specific and less toxic pharmacologic options. Based on the blueprint of non-canonical signaling in Drosophila, here, we review and rationalize current evidence about non-canonical Notch signaling. Our effort might inform Notch biologists developing new research avenues and clinicians seeking future treatment of Notch-dependent diseases.
Collapse
Affiliation(s)
- Victor Alfred
- IFOM, Istituto FIRC di Oncologia Molecolare at IFOM-IEO Campus, Milan, Italy
| | - Thomas Vaccari
- IFOM, Istituto FIRC di Oncologia Molecolare at IFOM-IEO Campus, Milan, Italy.
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
24
|
Stoten CL, Carlton JG. ESCRT-dependent control of membrane remodelling during cell division. Semin Cell Dev Biol 2017; 74:50-65. [PMID: 28843980 PMCID: PMC6015221 DOI: 10.1016/j.semcdb.2017.08.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/07/2017] [Accepted: 08/18/2017] [Indexed: 12/16/2022]
Abstract
The Endosomal Sorting Complex Required for Transport (ESCRT) proteins form an evolutionarily conserved membrane remodelling machinery. Identified originally for their role in cargo sorting and remodelling of endosomal membranes during yeast vacuolar sorting, an extensive body of work now implicates a sub-complex of this machinery (ESCRT-III), as a transplantable membrane fission machinery that is dispatched to various cellular locations to achieve a topologically unique membrane separation. Surprisingly, several ESCRT-III-regulated processes occur during cell division, when cells undergo a dramatic and co-ordinated remodelling of their membranes to allow the physical processes of division to occur. The ESCRT machinery functions in regeneration of the nuclear envelope during open mitosis and in the abscission phase of cytokinesis, where daughter cells are separated from each other in the last act of division. Roles for the ESCRT machinery in cell division are conserved as far back as Archaea, suggesting that the ancestral role of these proteins was as a membrane remodelling machinery that facilitated division and that was co-opted throughout evolution to perform a variety of other cell biological functions. Here, we will explore the function and regulation of the ESCRT machinery in cell division.
Collapse
|
25
|
Sadoul R, Laporte MH, Chassefeyre R, Chi KI, Goldberg Y, Chatellard C, Hemming FJ, Fraboulet S. The role of ESCRT during development and functioning of the nervous system. Semin Cell Dev Biol 2017; 74:40-49. [PMID: 28811263 DOI: 10.1016/j.semcdb.2017.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/21/2017] [Accepted: 08/04/2017] [Indexed: 12/12/2022]
Abstract
The endosomal sorting complex required for transport (ESCRT) is made of subcomplexes (ESCRT 0-III), crucial to membrane remodelling at endosomes, nuclear envelope and cell surface. ESCRT-III shapes membranes and in most cases cooperates with the ATPase VPS4 to mediate fission of membrane necks from the inside. The first ESCRT complexes mainly serve to catalyse the formation of ESCRT-III but can be bypassed by accessory proteins like the Alg-2 interacting protein-X (ALIX). In the nervous system, ALIX/ESCRT controls the survival of embryonic neural progenitors and later on the outgrowth and pruning of axons and dendrites, all necessary steps to establish a functional brain. In the adult brain, ESCRTs allow the endosomal turn over of synaptic vesicle proteins while stable ESCRT complexes might serve as scaffolds for the postsynaptic parts. The necessity of ESCRT for the harmonious function of the brain has its pathological counterpart, the mutations in CHMP2B of ESCRT-III giving rise to several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rémy Sadoul
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France.
| | - Marine H Laporte
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Romain Chassefeyre
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Kwang Il Chi
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Yves Goldberg
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Christine Chatellard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Fiona J Hemming
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Sandrine Fraboulet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| |
Collapse
|
26
|
Deshar R, Cho EB, Yoon SK, Yoon JB. CC2D1A and CC2D1B regulate degradation and signaling of EGFR and TLR4. Biochem Biophys Res Commun 2016; 480:280-287. [DOI: 10.1016/j.bbrc.2016.10.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/17/2016] [Indexed: 11/24/2022]
|
27
|
Tabata K, Arimoto M, Arakawa M, Nara A, Saito K, Omori H, Arai A, Ishikawa T, Konishi E, Suzuki R, Matsuura Y, Morita E. Unique Requirement for ESCRT Factors in Flavivirus Particle Formation on the Endoplasmic Reticulum. Cell Rep 2016; 16:2339-47. [PMID: 27545892 DOI: 10.1016/j.celrep.2016.07.068] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 11/28/2022] Open
Abstract
Flavivirus infection induces endoplasmic reticulum (ER) membrane rearrangements to generate a compartment for replication of the viral genome and assembly of viral particles. Using quantitative mass spectrometry, we identified several ESCRT (endosomal sorting complex required for transport) proteins that are recruited to sites of virus replication on the ER. Systematic small interfering RNA (siRNA) screening revealed that release of both dengue virus and Japanese encephalitis virus was dramatically decreased by single depletion of TSG101 or co-depletion of specific combinations of ESCRT-III proteins, resulting in ≥1,000-fold titer reductions. By contrast, release was unaffected by depletion of some core ESCRTs, including VPS4. Reintroduction of ESCRT proteins to siRNA-depleted cells revealed interactions among ESCRT proteins that are crucial for flavivirus budding. Electron-microscopy studies revealed that the CHMP2 and CHMP4 proteins function directly in membrane deformation at the ER. Thus, a unique and specific subset of ESCRT contributes to ER membrane biogenesis during flavivirus infection.
Collapse
Affiliation(s)
- Keisuke Tabata
- Laboratory of Viral Infection, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Masaru Arimoto
- Laboratory of Viral Infection, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Masashi Arakawa
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan
| | - Atsuki Nara
- Faculty of Bioscience, Nagahama Institute of Bio-science and Technology, Nagahama 526-0829, Japan
| | - Kazunobu Saito
- Core Instrumentation Facility, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Hiroko Omori
- Core Instrumentation Facility, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Arisa Arai
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan
| | - Tomohiro Ishikawa
- Department of Microbiology, Dokkyo Medical University School of Medicine, Tochigi 321-0293 Japan
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Eiji Morita
- Laboratory of Viral Infection, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan.
| |
Collapse
|
28
|
McMillan BJ, Tibbe C, Jeon H, Drabek AA, Klein T, Blacklow SC. Electrostatic Interactions between Elongated Monomers Drive Filamentation of Drosophila Shrub, a Metazoan ESCRT-III Protein. Cell Rep 2016; 16:1211-1217. [PMID: 27452459 PMCID: PMC4985235 DOI: 10.1016/j.celrep.2016.06.093] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/27/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) is a conserved protein complex that facilitates budding and fission of membranes. It executes a key step in many cellular events, including cytokinesis and multi-vesicular body formation. The ESCRT-III protein Shrub in flies, or its homologs in yeast (Snf7) or humans (CHMP4B), is a critical polymerizing component of ESCRT-III needed to effect membrane fission. We report the structural basis for polymerization of Shrub and define a minimal region required for filament formation. The X-ray structure of the Shrub core shows that individual monomers in the lattice interact in a staggered arrangement using complementary electrostatic surfaces. Mutations that disrupt interface salt bridges interfere with Shrub polymerization and function. Despite substantial sequence divergence and differences in packing interactions, the arrangement of Shrub subunits in the polymer resembles that of Snf7 and other family homologs, suggesting that this intermolecular packing mechanism is shared among ESCRT-III proteins.
Collapse
Affiliation(s)
- Brian J McMillan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Hyesung Jeon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrew A Drabek
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Klein
- Heinrich-Heine-University, Düsseldorf 40225, Germany
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
29
|
Gonçalves SA, Outeiro TF. Traffic jams and the complex role of α-Synuclein aggregation in Parkinson disease. Small GTPases 2016; 8:78-84. [PMID: 27314512 DOI: 10.1080/21541248.2016.1199191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A common pathological event among various neurodegenerative disorders (NDs) is the misfolding and aggregation of different proteins in the brain. This is thought to potentiate aberrant protein-protein interactions that culminate in the disruption of several biological processes and, ultimately, in neuronal cell loss. Although protein aggregates are a common hallmark in several disorders, the molecular pathways leading to their generation remain unclear. The misfolding and aggregation of α-Synuclein (aSyn) is the pathological hallmark of Parkinson disease (PD), the second most common age related ND. It has been postulated that oligomeric species of aSyn, rather than more mature aggregated forms of the protein, are the causative agents of cytotoxicity. In recent years, we have been investigating the molecular mechanisms underlying the initial steps of aSyn accumulation in living cells. Using an unbiased genome-wide lentiviral RNAi screen we identified trafficking and kinase genes as modulators of aSyn oligomerization, aggregation, and toxicity. Among those, Rab8b, Rab11a, Rab13 and Slp5 were found to promote the clearance of aSyn inclusions and reduce aSyn toxicity. Moreover, we found that endocytic recycling and secretion of aSyn was enhanced upon expression of Rab11a or Rab13 in cells accumulating aSyn inclusions. Altogether, our findings suggest specific trafficking steps may prove beneficial as targets for therapeutic intervention in synucleinopathies, and should be further investigated in other models.
Collapse
Affiliation(s)
- Susana A Gonçalves
- a CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Tiago Fleming Outeiro
- a CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal.,b Department of Neurodegeneration and Restorative Research , University Medical Center Göttingen , Göttingen , Germany.,c Max Planck Institute for Experimental Medicine , Göttingen , Germany
| |
Collapse
|
30
|
Gonçalves SA, Macedo D, Raquel H, Simões PD, Giorgini F, Ramalho JS, Barral DC, Ferreira Moita L, Outeiro TF. shRNA-Based Screen Identifies Endocytic Recycling Pathway Components That Act as Genetic Modifiers of Alpha-Synuclein Aggregation, Secretion and Toxicity. PLoS Genet 2016; 12:e1005995. [PMID: 27123591 PMCID: PMC4849646 DOI: 10.1371/journal.pgen.1005995] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/28/2016] [Indexed: 12/15/2022] Open
Abstract
Alpha-Synuclein (aSyn) misfolding and aggregation is common in several neurodegenerative diseases, including Parkinson's disease and dementia with Lewy bodies, which are known as synucleinopathies. Accumulating evidence suggests that secretion and cell-to-cell trafficking of pathological forms of aSyn may explain the typical patterns of disease progression. However, the molecular mechanisms controlling aSyn aggregation and spreading of pathology are still elusive. In order to obtain unbiased information about the molecular regulators of aSyn oligomerization, we performed a microscopy-based large-scale RNAi screen in living cells. Interestingly, we identified nine Rab GTPase and kinase genes that modulated aSyn aggregation, toxicity and levels. From those, Rab8b, Rab11a, Rab13 and Slp5 were able to promote the clearance of aSyn inclusions and rescue aSyn induced toxicity. Furthermore, we found that endocytic recycling and secretion of aSyn was enhanced upon Rab11a and Rab13 expression in cells accumulating aSyn inclusions. Overall, our study resulted in the identification of new molecular players involved in the aggregation, toxicity, and secretion of aSyn, opening novel avenues for our understanding of the molecular basis of synucleinopathies.
Collapse
Affiliation(s)
- Susana A. Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diana Macedo
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de lisboa, Oeiras, Portugal
| | | | - Pedro D. Simões
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| | - Flaviano Giorgini
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - José S. Ramalho
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Duarte C. Barral
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | | | - Tiago Fleming Outeiro
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
31
|
Drusenheimer N, Migdal B, Jäckel S, Tveriakhina L, Scheider K, Schulz K, Gröper J, Köhrer K, Klein T. The Mammalian Orthologs of Drosophila Lgd, CC2D1A and CC2D1B, Function in the Endocytic Pathway, but Their Individual Loss of Function Does Not Affect Notch Signalling. PLoS Genet 2015; 11:e1005749. [PMID: 26720614 PMCID: PMC4697852 DOI: 10.1371/journal.pgen.1005749] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/24/2015] [Indexed: 12/14/2022] Open
Abstract
CC2D1A and CC2D1B belong to the evolutionary conserved Lgd protein family with members in all multi-cellular animals. Several functions such as centrosomal cleavage, involvement in signalling pathways, immune response and synapse maturation have been described for CC2D1A. Moreover, the Drosophila melanogaster ortholog Lgd was shown to be involved in the endosomal trafficking of the Notch receptor and other transmembrane receptors and physically interacts with the ESCRT-III component Shrub/CHMP4. To determine if this function is conserved in mammals we generated and characterized Cc2d1a and Cc2d1b conditional knockout mice. While Cc2d1b deficient mice displayed no obvious phenotype, we found that Cc2d1a deficient mice as well as conditional mutants that lack CC2D1A only in the nervous system die shortly after birth due to respiratory distress. This finding confirms the suspicion that the breathing defect is caused by the central nervous system. However, an involvement in centrosomal function could not be confirmed in Cc2d1a deficient MEF cells. To analyse an influence on Notch signalling, we generated intestine specific Cc2d1a mutant mice. These mice did not display any alterations in goblet cell number, proliferating cell number or expression of the Notch reporter Hes1-emGFP, suggesting that CC2D1A is not required for Notch signalling. However, our EM analysis revealed that the average size of endosomes of Cc2d1a mutant cells, but not Cc2d1b mutant cells, is increased, indicating a defect in endosomal morphogenesis. We could show that CC2D1A and its interaction partner CHMP4B are localised on endosomes in MEF cells, when the activity of the endosomal protein VPS4 is reduced. This indicates that CC2D1A cycles between the cytosol and the endosomal membrane. Additionally, in rescue experiments in D. melanogaster, CC2D1A and CC2D1B were able to functionally replace Lgd. Altogether our data suggest a functional conservation of the Lgd protein family in the ESCRT-III mediated process in metazoans. The proteins of the Lgd/CC2D1 family are conserved in all multicellular animals. The Drosophila melanogaster ortholog Lgd is involved in the regulation of signalling receptor degradation via the endosomal pathway. Loss of lgd function causes ectopic ligand-independent activation of the Notch signalling pathway due to a defect in the endosomal pathway. For the mammalian proteins no endosomal function has been defined so far. Here, we asked whether the function of Lgd is conserved in mammals with the focus on the question whether its orthologs are also involved in the endosomal pathway and regulation of Notch pathway activity. Therefore, we generated and characterised Cc2d1a and Cc2d1b conditional knockout mice. We found that the loss of Cc2d1b does not lead to an obvious phenotype, while the known lethality of Cc2d1a deficient newborns is nervous system dependent. In experiments with MEFs isolated from knockout animals we provide evidence that both CC2D1 proteins are involved in the function of the ESCRT-III complex in a similar manner as Lgd in D. melanogaster. Moreover, we found that the loss of one CC2D1 protein is not sufficient to cause ectopic activation of Notch signalling.
Collapse
Affiliation(s)
- Nadja Drusenheimer
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
- * E-mail: (ND); (TK)
| | - Bernhard Migdal
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Sandra Jäckel
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Lena Tveriakhina
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Hannover, Germany
| | - Kristina Scheider
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Katharina Schulz
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Jieny Gröper
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Genomics and Transcriptomics Laboratory (GTL), Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Thomas Klein
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
- * E-mail: (ND); (TK)
| |
Collapse
|
32
|
Tang S, Henne WM, Borbat PP, Buchkovich NJ, Freed JH, Mao Y, Fromme JC, Emr SD. Structural basis for activation, assembly and membrane binding of ESCRT-III Snf7 filaments. eLife 2015; 4:e12548. [PMID: 26670543 PMCID: PMC4720517 DOI: 10.7554/elife.12548] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 12/13/2015] [Indexed: 12/14/2022] Open
Abstract
The endosomal sorting complexes required for transport (ESCRTs) constitute hetero-oligomeric machines that catalyze multiple topologically similar membrane-remodeling processes. Although ESCRT-III subunits polymerize into spirals, how individual ESCRT-III subunits are activated and assembled together into a membrane-deforming filament remains unknown. Here, we determine X-ray crystal structures of the most abundant ESCRT-III subunit Snf7 in its active conformation. Using pulsed dipolar electron spin resonance spectroscopy (PDS), we show that Snf7 activation requires a prominent conformational rearrangement to expose protein-membrane and protein-protein interfaces. This promotes the assembly of Snf7 arrays with ~30 Å periodicity into a membrane-sculpting filament. Using a combination of biochemical and genetic approaches, both in vitro and in vivo, we demonstrate that mutations on these protein interfaces halt Snf7 assembly and block ESCRT function. The architecture of the activated and membrane-bound Snf7 polymer provides crucial insights into the spatially unique ESCRT-III-mediated membrane remodeling.
Collapse
Affiliation(s)
- Shaogeng Tang
- Weill Institute of Cell and Molecular Biology, Cornell University, Ithaca, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - W Mike Henne
- Weill Institute of Cell and Molecular Biology, Cornell University, Ithaca, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Peter P Borbat
- National Biomedical Center for Advanced Electron Spin Resonance Technology, Cornell University, Ithaca, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, United States
| | - Nicholas J Buchkovich
- Weill Institute of Cell and Molecular Biology, Cornell University, Ithaca, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Jack H Freed
- National Biomedical Center for Advanced Electron Spin Resonance Technology, Cornell University, Ithaca, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, United States
| | - Yuxin Mao
- Weill Institute of Cell and Molecular Biology, Cornell University, Ithaca, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - J Christopher Fromme
- Weill Institute of Cell and Molecular Biology, Cornell University, Ithaca, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Scott D Emr
- Weill Institute of Cell and Molecular Biology, Cornell University, Ithaca, United States
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
33
|
Abstract
The endosomal sorting complexes required for transport (ESCRTs) collectively comprise a machinery that was first known for its function in the degradation of transmembrane proteins in the endocytic pathway of eukaryotic cells. Since their discovery, however, ESCRTs have been recognized as playing important roles at the plasma membrane, which appears to be the original site of function for the ESCRT machinery. This article reviews some of the major research findings that have shaped our current understanding of how the ESCRT machinery controls membrane dynamics and considers new roles for the ESCRT machinery that might be driven by these mechanisms.
Collapse
Affiliation(s)
- Greg Odorizzi
- Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, USA
| |
Collapse
|
34
|
Morawa KS, Schneider M, Klein T. Lgd regulates the activity of the BMP/Dpp signalling pathway during Drosophila oogenesis. Development 2015; 142:1325-35. [DOI: 10.1242/dev.112961] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The tumour suppressor gene lethal (2) giant discs (lgd) is involved in endosomal trafficking of transmembrane proteins in Drosophila. Loss of function results in the ligand-independent activation of the Notch pathway in all imaginal disc cells and follicle cells. Analysis of lgd loss of function has largely been restricted to imaginal discs and suggests that no other signalling pathway is affected. The devotion of Lgd to the Notch pathway was puzzling given that lgd loss of function also affects trafficking of components of other signalling pathways, such as the Dpp (a Drosophila BMP) pathway. Moreover, Lgd physically interacts with Shrub, a fundamental component of the ESCRT trafficking machinery, whose loss of function results in the activation of several signalling pathways. Here, we show that during oogenesis lgd loss of function causes ectopic activation of the Drosophila BMP signalling pathway. This activation occurs in somatic follicle cells as well as in germline cells. The activation in germline cells causes an extra round of division, producing egg chambers with 32 instead of 16 cells. Moreover, more germline stem cells were formed. The lgd mutant cells are defective in endosomal trafficking, causing an accumulation of the type I Dpp receptor Thickveins in maturing endosomes, which probably causes activation of the pathway. Taken together, these results show that lgd loss of function causes various effects among tissues and can lead to the activation of signalling pathways other than Notch. They further show that there is a role for the endosomal pathway during oogenesis.
Collapse
Affiliation(s)
- Kim Sara Morawa
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Universitätsstr.1, Düsseldorf 40225, Germany
| | - Markus Schneider
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Universitätsstr.1, Düsseldorf 40225, Germany
| | - Thomas Klein
- Institut für Genetik, Heinrich-Heine-Universität Düsseldorf, Universitätsstr.1, Düsseldorf 40225, Germany
| |
Collapse
|
35
|
Matias NR, Mathieu J, Huynh JR. Abscission is regulated by the ESCRT-III protein shrub in Drosophila germline stem cells. PLoS Genet 2015; 11:e1004653. [PMID: 25647097 PMCID: PMC4372032 DOI: 10.1371/journal.pgen.1004653] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 07/31/2014] [Indexed: 11/18/2022] Open
Abstract
Abscission is the final event of cytokinesis that leads to the physical separation of the two daughter cells. Recent technical advances have allowed a better understanding of the cellular and molecular events leading to abscission in isolated yeast or mammalian cells. However, how abscission is regulated in different cell types or in a developing organism remains poorly understood. Here, we characterized the function of the ESCRT-III protein Shrub during cytokinesis in germ cells undergoing a series of complete and incomplete divisions. We found that Shrub is required for complete abscission, and that levels of Shrub are critical for proper timing of abscission. Loss or gain of Shrub delays abscission in germline stem cells (GSCs), and leads to the formation of stem-cysts, where daughter cells share the same cytoplasm as the mother stem cell and cannot differentiate. In addition, our results indicate a negative regulation of Shrub by the Aurora B kinase during GSC abscission. Finally, we found that Lethal giant discs (lgd), known to be required for Shrub function in the endosomal pathway, also regulates the duration of abscission in GSCs. Abscission is the final step of cytokinesis which allows the physical separation of sister cells through the scission of a thin cytoplasmic bridge that links them at the end of mitosis. The duration of abscission varies depending on cell types, indicating that the event is developmentally regulated. Recently, we have identified two kinases, Aurora B and CycB/Cdk-1, which regulate the timing of abscission in germ cells and in mammalian cells. However, these kinases are upstream regulators and do not perform abscission per se. Here, we show that Shrub, a potential target of Aurora B and one of the most downstream effectors of abscission, is required for complete abscission in germline stem cells. In the absence of Shrub, the mother stem cell remains linked to its daughter cells, which then share the same cytoplasm and cannot differentiate. Loss of Shrub and Aurora B have opposite effects on abscission duration suggesting that Aurora B regulates negatively Shrub. We further show that Shrub acts together with its interactor Lethal giant disc to ensure proper abscission timing.
Collapse
Affiliation(s)
- Neuza Reis Matias
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
- CNRS UMR3215, Inserm U934, Paris, France
| | - Juliette Mathieu
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
- CNRS UMR3215, Inserm U934, Paris, France
- * E-mail: (JM); (JRH)
| | - Jean-René Huynh
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
- CNRS UMR3215, Inserm U934, Paris, France
- * E-mail: (JM); (JRH)
| |
Collapse
|
36
|
Shen QT, Schuh AL, Zheng Y, Quinney K, Wang L, Hanna M, Mitchell JC, Otegui MS, Ahlquist P, Cui Q, Audhya A. Structural analysis and modeling reveals new mechanisms governing ESCRT-III spiral filament assembly. ACTA ACUST UNITED AC 2014; 206:763-77. [PMID: 25202029 PMCID: PMC4164947 DOI: 10.1083/jcb.201403108] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cryo-EM and molecular dynamics simulations reveal unexpected flexibility in individual monomers and a stable interface between monomers in the spiral filaments formed by the ESCRT-III subunit Vps32/CHMP4B. The scission of biological membranes is facilitated by a variety of protein complexes that bind and manipulate lipid bilayers. ESCRT-III (endosomal sorting complex required for transport III) filaments mediate membrane scission during the ostensibly disparate processes of multivesicular endosome biogenesis, cytokinesis, and retroviral budding. However, mechanisms by which ESCRT-III subunits assemble into a polymer remain unknown. Using cryogenic electron microscopy (cryo-EM), we found that the full-length ESCRT-III subunit Vps32/CHMP4B spontaneously forms single-stranded spiral filaments. The resolution afforded by two-dimensional cryo-EM combined with molecular dynamics simulations revealed that individual Vps32/CHMP4B monomers within a filament are flexible and able to accommodate a range of bending angles. In contrast, the interface between monomers is stable and refractory to changes in conformation. We additionally found that the carboxyl terminus of Vps32/CHMP4B plays a key role in restricting the lateral association of filaments. Our findings highlight new mechanisms by which ESCRT-III filaments assemble to generate a unique polymer capable of membrane remodeling in multiple cellular contexts.
Collapse
Affiliation(s)
- Qing-Tao Shen
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Amber L Schuh
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Yuqing Zheng
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Kyle Quinney
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Lei Wang
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Michael Hanna
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Julie C Mitchell
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Marisa S Otegui
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Paul Ahlquist
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Qiang Cui
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706 Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| | - Anjon Audhya
- Department of Biomolecular Chemistry, School of Medicine and Public Health, Department of Botany, Department of Genetics, Department of Chemistry, Graduate Program in Biophysics, Department of Mathematics, Department of Biochemistry, Institute for Molecular Virology, Howard Hughes Medical Institute, and Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
37
|
Manzini MC, Xiong L, Shaheen R, Tambunan DE, Di Costanzo S, Mitisalis V, Tischfield DJ, Cinquino A, Ghaziuddin M, Christian M, Jiang Q, Laurent S, Nanjiani ZA, Rasheed S, Hill RS, Lizarraga SB, Gleason D, Sabbagh D, Salih MA, Alkuraya FS, Walsh CA. CC2D1A regulates human intellectual and social function as well as NF-κB signaling homeostasis. Cell Rep 2014; 8:647-55. [PMID: 25066123 PMCID: PMC4334362 DOI: 10.1016/j.celrep.2014.06.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/26/2014] [Accepted: 06/20/2014] [Indexed: 11/26/2022] Open
Abstract
Autism spectrum disorder (ASD) and intellectual disability (ID) are often comorbid, but the extent to which they share common genetic causes remains controversial. Here, we present two autosomal-recessive "founder" mutations in the CC2D1A gene causing fully penetrant cognitive phenotypes, including mild-to-severe ID, ASD, as well as seizures, suggesting shared developmental mechanisms. CC2D1A regulates multiple intracellular signaling pathways, and we found its strongest effect to be on the transcription factor nuclear factor κB (NF-κB). Cc2d1a gain and loss of function both increase activation of NF-κB, revealing a critical role of Cc2d1a in homeostatic control of intracellular signaling. Cc2d1a knockdown in neurons reduces dendritic complexity and increases NF-κB activity, and the effects of Cc2d1a depletion can be rescued by inhibiting NF-κB activity. Homeostatic regulation of neuronal signaling pathways provides a mechanism whereby common founder mutations could manifest diverse symptoms in different patients.
Collapse
Affiliation(s)
- M Chiara Manzini
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Lan Xiong
- Department of Psychiatry, Research Centre of Montreal Mental Health University Institute, University of Montreal, Montreal, QC H1N 3V2, Canada; University of Montreal Hospital Research Centre, Montreal, QC H2L 2W5, Canada
| | - Ranad Shaheen
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Dimira E Tambunan
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Stefania Di Costanzo
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Vanessa Mitisalis
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - David J Tischfield
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Antonella Cinquino
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Mohammed Ghaziuddin
- Department of Child and Adolescent Psychiatry, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Mehtab Christian
- University of Montreal Hospital Research Centre, Montreal, QC H2L 2W5, Canada
| | - Qin Jiang
- Department of Psychiatry, Research Centre of Montreal Mental Health University Institute, University of Montreal, Montreal, QC H1N 3V2, Canada
| | - Sandra Laurent
- University of Montreal Hospital Research Centre, Montreal, QC H2L 2W5, Canada
| | - Zohair A Nanjiani
- Ma Ayesha Memorial Centre, University of Karachi, Karachi 75350, Pakistan
| | | | - R Sean Hill
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Sofia B Lizarraga
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Danielle Gleason
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Diya Sabbagh
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Mustafa A Salih
- Division of Pediatric Neurology, Department of Pediatrics, King Saud University College of Medicine, Riyadh 11461, Saudi Arabia.
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia.
| | - Christopher A Walsh
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
38
|
Cashikar AG, Shim S, Roth R, Maldazys MR, Heuser JE, Hanson PI. Structure of cellular ESCRT-III spirals and their relationship to HIV budding. eLife 2014; 3. [PMID: 24878737 PMCID: PMC4073282 DOI: 10.7554/elife.02184] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/27/2014] [Indexed: 12/23/2022] Open
Abstract
The ESCRT machinery along with the AAA+ ATPase Vps4 drive membrane scission for trafficking into multivesicular bodies in the endocytic pathway and for the topologically related processes of viral budding and cytokinesis, but how they accomplish this remains unclear. Using deep-etch electron microscopy, we find that endogenous ESCRT-III filaments stabilized by depleting cells of Vps4 create uniform membrane-deforming conical spirals which are assemblies of specific ESCRT-III heteropolymers. To explore functional roles for ESCRT-III filaments, we examine HIV-1 Gag-mediated budding of virus-like particles and find that depleting Vps4 traps ESCRT-III filaments around nascent Gag assemblies. Interpolating between the observed structures suggests a new role for Vps4 in separating ESCRT-III from Gag or other cargo to allow centripetal growth of a neck constricting ESCRT-III spiral. DOI:http://dx.doi.org/10.7554/eLife.02184.001 Cells contain compartments called organelles that are enclosed within membranes similar to the plasma membrane that surrounds the cell itself. Cells police the proteins on their membranes and move old or damaged proteins into a type of organelle called an endosome. This involves the membrane folding in on itself to form a multivesicular body. The multivesicular bodies deliver their contents to organelles called lysosomes where the old proteins are destroyed. Although it is known that over 30 proteins are involved in the formation of multivesicular bodies, many aspects of how they operate are not well understood. Moreover, disruptions to this process contribute to a large number of diseases including forms of cancer and neurodegeneration. Importantly, the same proteins are hijacked by viruses such as HIV to help them escape from the cells they have infected. Most of the proteins involved in forming multivesicular bodies are part of the ESCRT (Endosomal Sorting Complex Required for Transport) system of proteins. A special set of these proteins—ESCRT-III—is thought to cut the membrane to release vesicles and viruses, as well as helping the membrane to deform. Previously, researchers have been unsure how the ESCRT-III complex works because it has a short lifespan and is too small to see on cellular membranes using standard techniques. Now Cashikar, Shim et al. have used a technique called deep-etch electron microscopy in combination with gene knockdown strategies to reveal the structure of the ESCRT-III complex inside cells. A protein called Vps4 is known to recycle ESCRT-III complexes, so Cashikar, Shim et al. studied cells in which the levels of Vps4 had been depleted in order to increase the lifespan of ESCRT-III complexes. In these cells filaments made of ESCRT-III complexes tended to form conical spirals that matched the size and shape of the vesicles and viruses ESCRT-III is thought to produce. ESCRT-III filaments also accumulated as rings around the molecules destined for incorporation into a vesicle or virus. This indicated a new role for Vps4: it separates ESCRT-III from the contents of the vesicle, leaving it free to form a spiral that drives release of the vesicle or virus from the cell. The next challenge will be to test the predictions of this model using techniques that can capture individual vesicle formation events in real time. Understanding the function of ESCRT-III in greater detail may suggest ways to manipulate this pathway to limit the replication of viruses or the degradation of membrane proteins. DOI:http://dx.doi.org/10.7554/eLife.02184.002
Collapse
Affiliation(s)
- Anil G Cashikar
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Soomin Shim
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Robyn Roth
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Michael R Maldazys
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - John E Heuser
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - Phyllis I Hanson
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
39
|
Schuh AL, Audhya A. The ESCRT machinery: from the plasma membrane to endosomes and back again. Crit Rev Biochem Mol Biol 2014; 49:242-61. [PMID: 24456136 DOI: 10.3109/10409238.2014.881777] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The manipulation and reorganization of lipid bilayers are required for diverse cellular processes, ranging from organelle biogenesis to cytokinetic abscission, and often involves transient membrane disruption. A set of membrane-associated proteins collectively known as the endosomal sorting complex required for transport (ESCRT) machinery has been implicated in membrane scission steps, which transform a single, continuous bilayer into two distinct bilayers, while simultaneously segregating cargo throughout the process. Components of the ESCRT pathway, which include 5 distinct protein complexes and an array of accessory factors, each serve discrete functions. This review focuses on the molecular mechanisms by which the ESCRT proteins facilitate cargo sequestration and membrane remodeling and highlights their unique roles in cellular homeostasis.
Collapse
Affiliation(s)
- Amber L Schuh
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health , Madison, WI , USA
| | | |
Collapse
|
40
|
Abstract
The endosomal sorting complexes required for transport (ESCRT) drive multivesicular body (MVB) biogenesis and cytokinetic abscission. Originally identified through genetics and cell biology, more recent work has begun to elucidate the molecular mechanisms of ESCRT-mediated membrane remodeling, with special focus on the ESCRT-III complex. In particular, several light and electron microscopic studies provide high-resolution imaging of ESCRT-III rings and spirals that purportedly drive MVB morphogenesis and abscission. These studies highlight unifying principles to ESCRT-III function, in particular: (1) the ordered assembly of the ESCRT-III monomers into a heteropolymer, (2) ESCRT-III as a dynamic complex, and (3) the role of the AAA ATPase Vps4 as a contributing factor in membrane scission. Mechanistic comparisons of ESCRT-III function in MVB morphogenesis and cytokinesis suggest common mechanisms in membrane remodeling.
Collapse
|
41
|
Abstract
The endosomal sorting complexes required for transport (ESCRT) pathway was initially defined in yeast genetic screens that identified the factors necessary to sort membrane proteins into intraluminal endosomal vesicles. Subsequent studies have revealed that the mammalian ESCRT pathway also functions in a series of other key cellular processes, including formation of extracellular microvesicles, enveloped virus budding, and the abscission stage of cytokinesis. The core ESCRT machinery comprises Bro1 family proteins and ESCRT-I, ESCRT-II, ESCRT-III, and VPS4 complexes. Site-specific adaptors recruit these soluble factors to assemble on different cellular membranes, where they carry out membrane fission reactions. ESCRT-III proteins form filaments that draw membranes together from the cytoplasmic face, and mechanistic models have been advanced to explain how ESCRT-III filaments and the VPS4 ATPase can work together to catalyze membrane fission.
Collapse
Affiliation(s)
- John McCullough
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112-5650, USA
| | | | | |
Collapse
|
42
|
Meng B, Lever AM. Wrapping up the bad news: HIV assembly and release. Retrovirology 2013; 10:5. [PMID: 23305486 PMCID: PMC3558412 DOI: 10.1186/1742-4690-10-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 12/21/2012] [Indexed: 02/02/2023] Open
Abstract
The late Nobel Laureate Sir Peter Medawar once memorably described viruses as ‘bad news wrapped in protein’. Virus assembly in HIV is a remarkably well coordinated process in which the virus achieves extracellular budding using primarily intracellular budding machinery and also the unusual phenomenon of export from the cell of an RNA. Recruitment of the ESCRT system by HIV is one of the best documented examples of the comprehensive way in which a virus hijacks a normal cellular process. This review is a summary of our current understanding of the budding process of HIV, from genomic RNA capture through budding and on to viral maturation, but centering on the proteins of the ESCRT pathway and highlighting some recent advances in our understanding of the cellular components involved and the complex interplay between the Gag protein and the genomic RNA.
Collapse
Affiliation(s)
- Bo Meng
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | |
Collapse
|
43
|
Effantin G, Dordor A, Sandrin V, Martinelli N, Sundquist WI, Schoehn G, Weissenhorn W. ESCRT-III CHMP2A and CHMP3 form variable helical polymers in vitro and act synergistically during HIV-1 budding. Cell Microbiol 2012; 15:213-26. [PMID: 23051622 DOI: 10.1111/cmi.12041] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/03/2012] [Accepted: 10/04/2012] [Indexed: 12/23/2022]
Abstract
The endosomal sorting complex required for transport-III (ESCRT-III) proteins are essential for budding of some enveloped viruses, for the formation of intraluminal vesicles at the endosome and for the abscission step of cytokinesis. ESCRT-III proteins form polymers that constrict membrane tubes, leading to fission. We have used electron cryomicroscopy to determine the molecular organization of pleiomorphic ESCRT-III CHMP2A-CHMP3 polymers. The three-dimensional reconstruction at 22 Å resolution reveals a helical organization of filaments of CHMP molecules organized in a head-to-tail fashion. Protease susceptibility experiments indicate that polymerization is achieved via conformational changes that increase the protomer stability. Combinatorial siRNA knockdown experiments indicate that CHMP3 contributes synergistically to HIV-1 budding, and the CHMP3 contribution is ~ 10-fold more pronounced in concert with CHMP2A than with CHMP2B. This is consistent with surface plasmon resonance affinity measurements that suggest sequential CHMP4B-CHMP3-CHMP2A recruitment while showing that both CHMP2A and CHMP2B interact with CHMP4B, in agreement with their redundant functions in HIV-1 budding. Our data thus indicate that the CHMP2A-CHMP3 polymer observed in vitro contributes to HIV-1 budding by assembling on CHMP4B polymers.
Collapse
Affiliation(s)
- Grégory Effantin
- Unit of Virus Host Cell Interactions, UMI 3265, Université Joseph Fourier-EMBL-CNRS, 6 rue Jules Horowitz, 38042 Grenoble Cedex 9, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Chen KR, Chang CH, Huang CY, Lin CY, Lin WY, Lo YC, Yang CY, Hsing EW, Chen LF, Shih SR, Shiau AL, Lei HY, Tan TH, Ling P. TBK1-associated protein in endolysosomes (TAPE)/CC2D1A is a key regulator linking RIG-I-like receptors to antiviral immunity. J Biol Chem 2012; 287:32216-21. [PMID: 22833682 PMCID: PMC3442552 DOI: 10.1074/jbc.c112.394346] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Indexed: 11/06/2022] Open
Abstract
Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are key RNA viral sensors for triggering antiviral immunity. The underlying mechanisms for RLRs to trigger antiviral immunity have yet to be explored. Here we report the identification of TAPE (TBK1-associated protein in endolysosomes) as a novel regulator of the RLR pathways. TAPE functionally and physically interacts with RIG-I, MDA5, and IPS-1 to activate the IFN-β promoter. TAPE knockdown impairs IFN-β activation induced by RLRs but not IPS-1. TAPE-deficient cells are defective in cytokine production upon RLR ligand stimulation. During RNA virus infection, TAPE knockdown or deficiency diminishes cytokine production and antiviral responses. Our data demonstrate a critical role for TAPE in linking RLRs to antiviral immunity.
Collapse
Affiliation(s)
| | | | - Ching-Yu Huang
- the Immunology Research Center, National Health Research Institutes, Zhunan 35053, Taiwan
| | | | - Wan-Ying Lin
- From the Departments of Microbiology and Immunology
| | - Yin-Chiu Lo
- From the Departments of Microbiology and Immunology
| | - Chia-Yu Yang
- the Immunology Research Center, National Health Research Institutes, Zhunan 35053, Taiwan
| | - En-Wei Hsing
- the Immunology Research Center, National Health Research Institutes, Zhunan 35053, Taiwan
- the Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | - Shin-Ru Shih
- the Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Ai-Li Shiau
- From the Departments of Microbiology and Immunology
- Institute of Basic Medical Sciences, and
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Huan-Yao Lei
- From the Departments of Microbiology and Immunology
- Institute of Basic Medical Sciences, and
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tse-Hua Tan
- the Immunology Research Center, National Health Research Institutes, Zhunan 35053, Taiwan
- the Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, and
| | - Pin Ling
- From the Departments of Microbiology and Immunology
- Institute of Basic Medical Sciences, and
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
45
|
Schneider M, Troost T, Grawe F, Martinez-Arias A, Klein T. Activation of Notch in lgd mutant cells requires the fusion of late endosomes with the lysosome. J Cell Sci 2012. [DOI: 10.1242/jcs.116590] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The tumorsuppressor Lethal (2) giant discs (Lgd) is a regulator of endosomal trafficking of the Notch signalling receptor as well as other transmembrane proteins in Drosophila. The loss of its function results in an uncontrolled ligand independent activation of of the Notch signalling receptor. Here, we further investigated the consequences of loss of lgd function and the requirements for the activation of Notch. We show that the activation of Notch in lgd cells is independent of Kuz and dependent on γ-secretase. We found that the lgd cells have a defect that delays degradation of transmembrane proteins, which are residents of the plasma membrane. Furthermore, our results show that the activation of Notch in lgd cells occurs in the lysosome. In contrast, the pathway is activated at an earlier phase in mutants of the gene that encodes the ESCRT-III component Shrub, which is an interaction partner of Lgd. We further show that activation of Notch appears to be a general consequence of loss of lgd function. In addition, we describe the EM analysis of lgd cells, which revealed that lgd cells contain enlarged MVBs. The presented results further elucidate the mechanism of uncontrolled Notch activation upon derailed endocytosis.
Collapse
|