1
|
Reshetniak S, Bogaciu CA, Bonn S, Brose N, Cooper BH, D'Este E, Fauth M, Fernández-Busnadiego R, Fiosins M, Fischer A, Georgiev SV, Jakobs S, Klumpp S, Köster S, Lange F, Lipstein N, Macarrón-Palacios V, Milovanovic D, Moser T, Müller M, Opazo F, Outeiro TF, Pape C, Priesemann V, Rehling P, Salditt T, Schlüter O, Simeth N, Steinem C, Tchumatchenko T, Tetzlaff C, Tirard M, Urlaub H, Wichmann C, Wolf F, Rizzoli SO. The synaptic vesicle cluster as a controller of pre- and postsynaptic structure and function. J Physiol 2024. [PMID: 39367860 DOI: 10.1113/jp286400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024] Open
Abstract
The synaptic vesicle cluster (SVC) is an essential component of chemical synapses, which provides neurotransmitter-loaded vesicles during synaptic activity, at the same time as also controlling the local concentrations of numerous exo- and endocytosis cofactors. In addition, the SVC hosts molecules that participate in other aspects of synaptic function, from cytoskeletal components to adhesion proteins, and affects the location and function of organelles such as mitochondria and the endoplasmic reticulum. We argue here that these features extend the functional involvement of the SVC in synapse formation, signalling and plasticity, as well as synapse stabilization and metabolism. We also propose that changes in the size of the SVC coalesce with changes in the postsynaptic compartment, supporting the interplay between pre- and postsynaptic dynamics. Thereby, the SVC could be seen as an 'all-in-one' regulator of synaptic structure and function, which should be investigated in more detail, to reveal molecular mechanisms that control synaptic function and heterogeneity.
Collapse
Affiliation(s)
- Sofiia Reshetniak
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Cristian A Bogaciu
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Elisa D'Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Michael Fauth
- Georg-August-University Göttingen, Faculty of Physics, Institute for the Dynamics of Complex Systems, Friedrich-Hund-Platz 1, Göttingen, Germany
| | - Rubén Fernández-Busnadiego
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Maksims Fiosins
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - André Fischer
- German Center for Neurodegenerative Diseases, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Svilen V Georgiev
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Jakobs
- Research Group Structure and Dynamics of Mitochondria, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Stefan Klumpp
- Theoretical Biophysics Group, Institute for the Dynamics of Complex Systems, Georg-August University Göttingen, Göttingen, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Felix Lange
- Research Group Structure and Dynamics of Mitochondria, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Noa Lipstein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Marcus Müller
- Institute for Theoretical Physics, Georg-August University Göttingen, Göttingen, Germany
| | - Felipe Opazo
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Constantin Pape
- Institute of Computer Science, Georg-August University Göttingen, Göttingen, Germany
| | - Viola Priesemann
- Georg-August-University Göttingen, Faculty of Physics, Institute for the Dynamics of Complex Systems, Friedrich-Hund-Platz 1, Göttingen, Germany
- Max-Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institute for X-Ray Physics, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Oliver Schlüter
- Clinic for Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nadja Simeth
- Institute of Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
| | - Christian Tetzlaff
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Marilyn Tirard
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Carolin Wichmann
- Institute for Auditory Neuroscience University Medical Center Göttingen, Göttingen, Germany
- Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Fred Wolf
- Max-Planck-Institute for Dynamics and Self-Organization, 37077 Göttingen and Institute for Dynamics of Biological Networks, Georg-August University Göttingen, Göttingen, Germany
| | - Silvio O Rizzoli
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
Shanley HT, Taki AC, Nguyen N, Wang T, Byrne JJ, Ang CS, Leeming MG, Williamson N, Chang BCH, Jabbar A, Sleebs BE, Gasser RB. Comparative structure activity and target exploration of 1,2-diphenylethynes in Haemonchus contortus and Caenorhabditis elegans. Int J Parasitol Drugs Drug Resist 2024; 25:100534. [PMID: 38554597 PMCID: PMC10992699 DOI: 10.1016/j.ijpddr.2024.100534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 04/01/2024]
Abstract
Infections and diseases caused by parasitic nematodes have a major adverse impact on the health and productivity of animals and humans worldwide. The control of these parasites often relies heavily on the treatment with commercially available chemical compounds (anthelmintics). However, the excessive or uncontrolled use of these compounds in livestock animals has led to major challenges linked to drug resistance in nematodes. Therefore, there is a need to develop new anthelmintics with novel mechanism(s) of action. Recently, we identified a small molecule, designated UMW-9729, with nematocidal activity against the free-living model organism Caenorhabditis elegans. Here, we evaluated UMW-9729's potential as an anthelmintic in a structure-activity relationship (SAR) study in C. elegans and the highly pathogenic, blood-feeding Haemonchus contortus (barber's pole worm), and explored the compound-target relationship using thermal proteome profiling (TPP). First, we synthesised and tested 25 analogues of UMW-9729 for their nematocidal activity in both H. contortus (larvae and adults) and C. elegans (young adults), establishing a preliminary nematocidal pharmacophore for both species. We identified several compounds with marked activity against either H. contortus or C. elegans which had greater efficacy than UMW-9729, and found a significant divergence in compound bioactivity between these two nematode species. We also identified a UMW-9729 analogue, designated 25, that moderately inhibited the motility of adult female H. contortus in vitro. Subsequently, we inferred three H. contortus proteins (HCON_00134350, HCON_00021470 and HCON_00099760) and five C. elegans proteins (F30A10.9, F15B9.8, B0361.6, DNC-4 and UNC-11) that interacted directly with UMW-9729; however, no conserved protein target was shared between the two nematode species. Future work aims to extend the SAR investigation in these and other parasitic nematode species, and validate individual proteins identified here as possible targets of UMW-9729. Overall, the present study evaluates this anthelmintic candidate and highlights some challenges associated with early anthelmintic investigation.
Collapse
Affiliation(s)
- Harrison T Shanley
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Aya C Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Nghi Nguyen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Joseph J Byrne
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Bill C H Chang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Brad E Sleebs
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
3
|
Moreira P, Papatheodorou P, Deng S, Gopal S, Handley A, Powell DR, Pocock R. Nuclear factor Y is a pervasive regulator of neuronal gene expression. Cell Rep 2023; 42:113582. [PMID: 38096055 DOI: 10.1016/j.celrep.2023.113582] [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: 02/12/2023] [Revised: 10/12/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Nervous system function relies on the establishment of complex gene expression programs that provide neuron-type-specific and core pan-neuronal features. These complementary regulatory paradigms are controlled by terminal selector and parallel-acting transcription factors (TFs), respectively. Here, we identify the nuclear factor Y (NF-Y) TF as a pervasive direct and indirect regulator of both neuron-type-specific and pan-neuronal gene expression. Mapping global NF-Y targets reveals direct binding to the cis-regulatory regions of pan-neuronal genes and terminal selector TFs. We show that NFYA-1 controls pan-neuronal gene expression directly through binding to CCAAT boxes in target gene promoters and indirectly by regulating the expression of terminal selector TFs. Further, we find that NFYA-1 regulation of neuronal gene expression is important for neuronal activity and motor function. Thus, our research sheds light on how global neuronal gene expression programs are buffered through direct and indirect regulatory mechanisms.
Collapse
Affiliation(s)
- Pedro Moreira
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Paul Papatheodorou
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Shuer Deng
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Sandeep Gopal
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Ava Handley
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - David R Powell
- Bioinformatics Platform, Monash University, Melbourne, VIC 3800, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
4
|
Petratou D, Gjikolaj M, Kaulich E, Schafer W, Tavernarakis N. A proton-inhibited DEG/ENaC ion channel maintains neuronal ionstasis and promotes neuronal survival under stress. iScience 2023; 26:107117. [PMID: 37416472 PMCID: PMC10320524 DOI: 10.1016/j.isci.2023.107117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/28/2023] [Accepted: 06/09/2023] [Indexed: 07/08/2023] Open
Abstract
The nervous system participates in the initiation and modulation of systemic stress. Ionstasis is of utmost importance for neuronal function. Imbalance in neuronal sodium homeostasis is associated with pathologies of the nervous system. However, the effects of stress on neuronal Na+ homeostasis, excitability, and survival remain unclear. We report that the DEG/ENaC family member DEL-4 assembles into a proton-inactivated sodium channel. DEL-4 operates at the neuronal membrane and synapse to modulate Caenorhabditis elegans locomotion. Heat stress and starvation alter DEL-4 expression, which in turn alters the expression and activity of key stress-response transcription factors and triggers appropriate motor adaptations. Similar to heat stress and starvation, DEL-4 deficiency causes hyperpolarization of dopaminergic neurons and affects neurotransmission. Using humanized models of neurodegenerative diseases in C. elegans, we showed that DEL-4 promotes neuronal survival. Our findings provide insights into the molecular mechanisms by which sodium channels promote neuronal function and adaptation under stress.
Collapse
Affiliation(s)
- Dionysia Petratou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, 70013 Crete, Greece
- Department of Basic Sciences, Medical School, University of Crete, Heraklion, 71003 Crete, Greece
| | - Martha Gjikolaj
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, 70013 Crete, Greece
- Department of Basic Sciences, Medical School, University of Crete, Heraklion, 71003 Crete, Greece
| | - Eva Kaulich
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, CB2 0QH Cambridge, UK
| | - William Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, CB2 0QH Cambridge, UK
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, 70013 Crete, Greece
- Department of Basic Sciences, Medical School, University of Crete, Heraklion, 71003 Crete, Greece
| |
Collapse
|
5
|
Krishnan S, Klingauf J. The readily retrievable pool of synaptic vesicles. Biol Chem 2023; 404:385-397. [PMID: 36867726 DOI: 10.1515/hsz-2022-0298] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023]
Abstract
In the CNS communication between neurons occurs at synapses by secretion of neurotransmitter via exocytosis of synaptic vesicles (SVs) at the active zone. Given the limited number of SVs in presynaptic boutons a fast and efficient recycling of exocytosed membrane and proteins by triggered compensatory endocytosis is required to maintain neurotransmission. Thus, pre-synapses feature a unique tight coupling of exo- and endocytosis in time and space resulting in the reformation of SVs with uniform morphology and well-defined molecular composition. This rapid response requires early stages of endocytosis at the peri-active zone to be well choreographed to ensure reformation of SVs with high fidelity. The pre-synapse can address this challenge by a specialized membrane microcompartment, where a pre-sorted and pre-assembled readily retrievable pool (RRetP) of endocytic membrane patches is formed, consisting of the vesicle cargo, presumably bound within a nucleated Clathrin and adaptor complex. This review considers evidence for the RRetP microcompartment to be the primary organizer of presynaptic triggered compensatory endocytosis.
Collapse
Affiliation(s)
- Sai Krishnan
- Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch Strasse 31, D-48149, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch Strasse 31, D-48149, Münster, Germany.,Center for Soft Nanoscience, Busso-Peus Strasse 10, D-48149, Münster, Germany
| |
Collapse
|
6
|
Ando K, Nagaraj S, Küçükali F, de Fisenne MA, Kosa AC, Doeraene E, Lopez Gutierrez L, Brion JP, Leroy K. PICALM and Alzheimer's Disease: An Update and Perspectives. Cells 2022; 11:3994. [PMID: 36552756 PMCID: PMC9776874 DOI: 10.3390/cells11243994] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified the PICALM (Phosphatidylinositol binding clathrin-assembly protein) gene as the most significant genetic susceptibility locus after APOE and BIN1. PICALM is a clathrin-adaptor protein that plays a critical role in clathrin-mediated endocytosis and autophagy. Since the effects of genetic variants of PICALM as AD-susceptibility loci have been confirmed by independent genetic studies in several distinct cohorts, there has been a number of in vitro and in vivo studies attempting to elucidate the underlying mechanism by which PICALM modulates AD risk. While differential modulation of APP processing and Aβ transcytosis by PICALM has been reported, significant effects of PICALM modulation of tau pathology progression have also been evidenced in Alzheimer's disease models. In this review, we summarize the current knowledge about PICALM, its physiological functions, genetic variants, post-translational modifications and relevance to AD pathogenesis.
Collapse
Affiliation(s)
- Kunie Ando
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Siranjeevi Nagaraj
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Fahri Küçükali
- Complex Genetics of Alzheimer’s Disease Group, VIB Center for Molecular Neurology, VIB Antwerp, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium
| | - Marie-Ange de Fisenne
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Andreea-Claudia Kosa
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Emilie Doeraene
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Lidia Lopez Gutierrez
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| |
Collapse
|
7
|
Xu H, Chang F, Jain S, Heller BA, Han X, Liu Y, Edwards RH. SNX5 targets a monoamine transporter to the TGN for assembly into dense core vesicles by AP-3. J Cell Biol 2022; 221:e202106083. [PMID: 35426896 PMCID: PMC9016777 DOI: 10.1083/jcb.202106083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/06/2021] [Accepted: 02/16/2022] [Indexed: 11/22/2022] Open
Abstract
The time course of signaling by peptide hormones, neural peptides, and other neuromodulators depends on their storage inside dense core vesicles (DCVs). Adaptor protein 3 (AP-3) assembles the membrane proteins that confer regulated release of DCVs and is thought to promote their trafficking from endosomes directly to maturing DCVs. We now find that regulated monoamine release from DCVs requires sorting nexin 5 (SNX5). Loss of SNX5 disrupts trafficking of the vesicular monoamine transporter (VMAT) to DCVs. The mechanism involves a role for SNX5 in retrograde transport of VMAT from endosomes to the TGN. However, this role for SNX5 conflicts with the proposed function of AP-3 in trafficking from endosomes directly to DCVs. We now identify a transient role for AP-3 at the TGN, where it associates with DCV cargo. Thus, retrograde transport from endosomes by SNX5 enables DCV assembly at the TGN by AP-3, resolving the apparent antagonism. A novel role for AP-3 at the TGN has implications for other organelles that also depend on this adaptor.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Fei Chang
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Shweta Jain
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| | - Bradley Austin Heller
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| | - Xu Han
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yongjian Liu
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Departments of Pharmacology and Biological Chemistry, University of Pittsburgh, Pittsburgh, PA
| | - Robert H. Edwards
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
8
|
Aniento F, Sánchez de Medina Hernández V, Dagdas Y, Rojas-Pierce M, Russinova E. Molecular mechanisms of endomembrane trafficking in plants. THE PLANT CELL 2022; 34:146-173. [PMID: 34550393 PMCID: PMC8773984 DOI: 10.1093/plcell/koab235] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/12/2021] [Indexed: 05/10/2023]
Abstract
Endomembrane trafficking is essential for all eukaryotic cells. The best-characterized membrane trafficking organelles include the endoplasmic reticulum (ER), Golgi apparatus, early and recycling endosomes, multivesicular body, or late endosome, lysosome/vacuole, and plasma membrane. Although historically plants have given rise to cell biology, our understanding of membrane trafficking has mainly been shaped by the much more studied mammalian and yeast models. Whereas organelles and major protein families that regulate endomembrane trafficking are largely conserved across all eukaryotes, exciting variations are emerging from advances in plant cell biology research. In this review, we summarize the current state of knowledge on plant endomembrane trafficking, with a focus on four distinct trafficking pathways: ER-to-Golgi transport, endocytosis, trans-Golgi network-to-vacuole transport, and autophagy. We acknowledge the conservation and commonalities in the trafficking machinery across species, with emphasis on diversity and plant-specific features. Understanding the function of organelles and the trafficking machinery currently nonexistent in well-known model organisms will provide great opportunities to acquire new insights into the fundamental cellular process of membrane trafficking.
Collapse
Affiliation(s)
| | - Víctor Sánchez de Medina Hernández
- Gregor Mendel Institute, Austrian Academy of Sciences, Vienna BioCenter, 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, A-1030, Vienna, Austria
| | | | | | | |
Collapse
|
9
|
Abstract
Axonal transport is an essential component of neuronal function. Several neurodegenerative disorders have been associated with defects in cargo transport. Thus, studying axonal transport is important to understand such disorders. Live imaging of fluorescently labeled cargo is a prevailing technique to study properties of axonal transport. C. elegans is both transparent and genetically amenable, making it an excellent model system to study axonal transport. In this chapter, we describe protocols to live image several neuronal cargo in vivo in C. elegans neurons.
Collapse
Affiliation(s)
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
10
|
Cousin MA. Synaptophysin-dependent synaptobrevin-2 trafficking at the presynapse-Mechanism and function. J Neurochem 2021; 159:78-89. [PMID: 34468992 DOI: 10.1111/jnc.15499] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/30/2022]
Abstract
Synaptobrevin-2 (Syb2) is a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) that is essential for neurotransmitter release. It is the most numerous protein on a synaptic vesicle (SV) and drives SV fusion via interactions with its cognate SNARE partners on the presynaptic plasma membrane. Synaptophysin (Syp) is the second most abundant protein on SVs; however, in contrast to Syb2, it has no obligatory role in neurotransmission. Syp interacts with Syb2 on SVs, and the molecular nature of its interaction with Syb2 and its physiological role has been debated for decades. However, recent studies have revealed that the sole physiological role of Syp at the presynapse is to ensure the efficient retrieval of Syb2 during SV endocytosis. In this review, current theories surrounding the role of Syp in Syb2 trafficking will be discussed, in addition to the debate regarding the molecular nature of their interaction. A unifying model is presented that describes how Syp controls Syb2 function as part of an integrated mechanism involving key molecular players such as intersectin-1 and AP180/CALM. Finally, key future questions surrounding the role of Syp-dependent Syb2 trafficking will be posed, with respect to brain function in health and disease.
Collapse
Affiliation(s)
- Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, Scotland, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
11
|
Sauvola CW, Littleton JT. SNARE Regulatory Proteins in Synaptic Vesicle Fusion and Recycling. Front Mol Neurosci 2021; 14:733138. [PMID: 34421538 PMCID: PMC8377282 DOI: 10.3389/fnmol.2021.733138] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Membrane fusion is a universal feature of eukaryotic protein trafficking and is mediated by the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) family. SNARE proteins embedded in opposing membranes spontaneously assemble to drive membrane fusion and cargo exchange in vitro. Evolution has generated a diverse complement of SNARE regulatory proteins (SRPs) that ensure membrane fusion occurs at the right time and place in vivo. While a core set of SNAREs and SRPs are common to all eukaryotic cells, a specialized set of SRPs within neurons confer additional regulation to synaptic vesicle (SV) fusion. Neuronal communication is characterized by precise spatial and temporal control of SNARE dynamics within presynaptic subdomains specialized for neurotransmitter release. Action potential-elicited Ca2+ influx at these release sites triggers zippering of SNAREs embedded in the SV and plasma membrane to drive bilayer fusion and release of neurotransmitters that activate downstream targets. Here we discuss current models for how SRPs regulate SNARE dynamics and presynaptic output, emphasizing invertebrate genetic findings that advanced our understanding of SRP regulation of SV cycling.
Collapse
Affiliation(s)
- Chad W Sauvola
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
12
|
Gallegos M, Hermes A, Patra D. Creation of a functional unc-11/PICALM GFP knock-in by CRISPR. MICROPUBLICATION BIOLOGY 2021; 2021:10.17912/micropub.biology.000389. [PMID: 33937722 PMCID: PMC8082290 DOI: 10.17912/micropub.biology.000389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
unc-11 is the only C. elegans ortholog of mammalian PICALM/AP180, paralogs that play an important role in Clathrin-Mediated Endocytosis (CME) and the recycling of a subset of SNAREs, the vesicle-associated membrane proteins (VAMPs). In this publication we report the creation of a new unc-11 allele that is endogenously-tagged with GFP just upstream of the stop codon. Moreover, we demonstrate that the UNC-11::GFP fusion protein functions like wild type with an expression pattern similar to UNC-11 antibody staining described previously.
Collapse
Affiliation(s)
- Maria Gallegos
- California State University, East Bay,
Correspondence to: Maria Gallegos ()
| | | | | |
Collapse
|
13
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
14
|
Luth ES, Hodul M, Rennich BJ, Riccio C, Hofer J, Markoja K, Juo P. VER/VEGF receptors regulate AMPA receptor surface levels and glutamatergic behavior. PLoS Genet 2021; 17:e1009375. [PMID: 33561120 PMCID: PMC7899335 DOI: 10.1371/journal.pgen.1009375] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 02/22/2021] [Accepted: 01/22/2021] [Indexed: 01/09/2023] Open
Abstract
Several intracellular trafficking pathways contribute to the regulation of AMPA receptor (AMPAR) levels at synapses and the control of synaptic strength. While much has been learned about these intracellular trafficking pathways, a major challenge is to understand how extracellular factors, such as growth factors, neuropeptides and hormones, impinge on specific AMPAR trafficking pathways to alter synaptic function and behavior. Here, we identify the secreted ligand PVF-1 and its cognate VEGF receptor homologs, VER-1 and VER-4, as regulators of glutamate signaling in C. elegans. Loss of function mutations in ver-1, ver-4, or pvf-1, result in decreased cell surface levels of the AMPAR GLR-1 and defects in glutamatergic behavior. Rescue experiments indicate that PVF-1 is expressed and released from muscle, whereas the VERs function in GLR-1-expressing neurons to regulate surface levels of GLR-1 and glutamatergic behavior. Additionally, ver-4 is unable to rescue glutamatergic behavior in the absence of pvf-1, suggesting that VER function requires endogenous PVF-1. Inducible expression of a pvf-1 rescuing transgene suggests that PVF-1 can function in the mature nervous system to regulate GLR-1 signaling. Genetic double mutant analysis suggests that the VERs act together with the VPS-35/retromer recycling complex to promote cell surface levels of GLR-1. Our data support a genetic model whereby PVF-1/VER signaling acts with retromer to promote recycling and cell surface levels of GLR-1 to control behavior. Sensation, behavior, and cognition all depend on the proper function of neuronal connections called synapses. Synapses that use the neurotransmitter glutamate to signal between nerve cells are the most abundant type in our brain. Presynaptic neurons release glutamate, which activates glutamate receptors on postsynaptic neurons. Dysfunction of glutamate synapses leads to several neurological disorders, and changing their strength–in part by altering glutamate receptors numbers on the surface of the postsynaptic cell—provides the cellular basis of learning and memory. Much remains to be learned about how factors released from other cell types affects synaptic communication. We took advantage of light-activated molecular switches engineered into specific sensory neurons of C. elegans worms to trigger a behavioral reflex that depends on glutamate synapses. Using this behavior, we identified proteins called VER-1 and VER-4 as important for glutamate synapse function. We found that worms missing these VER proteins or their activator PVF-1 have reduced levels of glutamate receptors at the postsynaptic surface and defects in glutamate-dependent behaviors. Our results suggest that inter-tissue cross-talk between muscle PVF-1 and neuronal VERs is important for controlling the number of glutamate receptors at the cell surface, robust neuronal communication and behavioral responses.
Collapse
Affiliation(s)
- Eric S. Luth
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biology, Simmons University, Boston, Massachusetts, United States of America
| | - Molly Hodul
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Bethany J. Rennich
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Carmino Riccio
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Julia Hofer
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Kaitlin Markoja
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Peter Juo
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
15
|
Treinin M, Jin Y. Cholinergic transmission in C. elegans: Functions, diversity, and maturation of ACh-activated ion channels. J Neurochem 2020; 158:1274-1291. [PMID: 32869293 DOI: 10.1111/jnc.15164] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Acetylcholine is an abundant neurotransmitter in all animals. Effects of acetylcholine are excitatory, inhibitory, or modulatory depending on the receptor and cell type. Research using the nematode C. elegans has made ground-breaking contributions to the mechanistic understanding of cholinergic transmission. Powerful genetic screens for behavioral mutants or for responses to pharmacological reagents identified the core cellular machinery for synaptic transmission. Pharmacological reagents that perturb acetylcholine-mediated processes led to the discovery and also uncovered the composition and regulators of acetylcholine-activated channels and receptors. From a combination of electrophysiological and molecular cellular studies, we have gained a profound understanding of cholinergic signaling at the levels of synapses, neural circuits, and animal behaviors. This review will begin with a historical overview, then cover in-depth current knowledge on acetylcholine-activated ionotropic receptors, mechanisms regulating their functional expression and their functions in regulating locomotion.
Collapse
Affiliation(s)
- Millet Treinin
- Department of Medical Neurobiology, Hadassah Medical school - Hebrew University, Jerusalem, Israel
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Pimentel-Acosta CA, Ramírez-Salcedo J, Morales-Serna FN, Fajer-Ávila EJ, Chávez-Sánchez C, Lara HH, García-Gasca A. Molecular Effects of Silver Nanoparticles on Monogenean Parasites: Lessons from Caenorhabditis elegans. Int J Mol Sci 2020; 21:ijms21165889. [PMID: 32824343 PMCID: PMC7460582 DOI: 10.3390/ijms21165889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022] Open
Abstract
The mechanisms of action of silver nanoparticles (AgNPs) in monogenean parasites of the genus Cichlidogyrus were investigated through a microarray hybridization approach using genomic information from the nematode Caenorhabditis elegans. The effects of two concentrations of AgNPs were explored, low (6 µg/L Ag) and high (36 µg/L Ag). Microarray analysis revealed that both concentrations of AgNPs activated similar biological processes, although by different mechanisms. Expression profiles included genes involved in detoxification, neurotoxicity, modulation of cell signaling, reproduction, embryonic development, and tegument organization as the main biological processes dysregulated by AgNPs. Two important processes (DNA damage and cell death) were mostly activated in parasites exposed to the lower concentration of AgNPs. To our knowledge, this is the first study providing information on the sub-cellular and molecular effects of exposure to AgNPs in metazoan parasites of fish.
Collapse
Affiliation(s)
- Citlalic A. Pimentel-Acosta
- Centro de Investigación en Alimentación y Desarrollo, Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán, Sinaloa 82112, Mexico; (C.A.P.-A.); (F.N.M.-S.); (E.J.F.-Á.); (C.C.-S.)
| | - Jorge Ramírez-Salcedo
- Unidad de Microarreglos, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Mexico City 04510, Mexico;
| | - Francisco Neptalí Morales-Serna
- Centro de Investigación en Alimentación y Desarrollo, Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán, Sinaloa 82112, Mexico; (C.A.P.-A.); (F.N.M.-S.); (E.J.F.-Á.); (C.C.-S.)
- CONACYT, Centro de Investigación en Alimentación y Desarrollo, Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán, Sinaloa 82112, Mexico
| | - Emma J. Fajer-Ávila
- Centro de Investigación en Alimentación y Desarrollo, Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán, Sinaloa 82112, Mexico; (C.A.P.-A.); (F.N.M.-S.); (E.J.F.-Á.); (C.C.-S.)
| | - Cristina Chávez-Sánchez
- Centro de Investigación en Alimentación y Desarrollo, Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán, Sinaloa 82112, Mexico; (C.A.P.-A.); (F.N.M.-S.); (E.J.F.-Á.); (C.C.-S.)
| | - Humberto H. Lara
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA;
| | - Alejandra García-Gasca
- Centro de Investigación en Alimentación y Desarrollo, Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán, Sinaloa 82112, Mexico; (C.A.P.-A.); (F.N.M.-S.); (E.J.F.-Á.); (C.C.-S.)
- Correspondence: ; Tel.: +52-66-9989-8700
| |
Collapse
|
17
|
Téllez-Arreola JL, Silva M, Martínez-Torres A. MCTP-1 modulates neurotransmitter release in C. elegans. Mol Cell Neurosci 2020; 107:103528. [PMID: 32650044 DOI: 10.1016/j.mcn.2020.103528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 11/30/2022] Open
Abstract
Multiple C2 and Transmembrane Domain Proteins (MCTPs) are putative calcium sensors. Proteins that contain C2 domains play essential roles in membrane trafficking and exocytosis; however, MCTPs functions in neurotransmitter release are not known. Here we report that in C. elegans mctp-1 is under the control of two promoters - one active in the nervous system and the second in the spermatheca. We generated and characterized a loss of function amt1 mutant and compared it to a previously published loss of function mutant (av112). Loss of mctp-1 function causes defects in egg-laying, crawling velocity, and thrashing rates. Both amt1 and av112 mutants are hyposensitive to the acetylcholinesterase blocker aldicarb, suggesting that MCTP-1 may play a role in synaptic vesicle release.
Collapse
Affiliation(s)
- José Luis Téllez-Arreola
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, C.P. 76215 Juriquilla, Querétaro, México; School of Biological Sciences, University of Utah, Salt Lake City, United States
| | - Malan Silva
- School of Biological Sciences, University of Utah, Salt Lake City, United States
| | - Ataúlfo Martínez-Torres
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, C.P. 76215 Juriquilla, Querétaro, México.
| |
Collapse
|
18
|
The Synaptic Vesicle Cycle Revisited: New Insights into the Modes and Mechanisms. J Neurosci 2020; 39:8209-8216. [PMID: 31619489 DOI: 10.1523/jneurosci.1158-19.2019] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 02/01/2023] Open
Abstract
Neurotransmission is sustained by endocytosis and refilling of synaptic vesicles (SVs) locally within the presynapse. Until recently, a consensus formed that after exocytosis, SVs are recovered by either fusion pore closure (kiss-and-run) or clathrin-mediated endocytosis directly from the plasma membrane. However, recent data have revealed that SV formation is more complex than previously envisaged. For example, two additional recycling pathways have been discovered, ultrafast endocytosis and activity-dependent bulk endocytosis, in which SVs are regenerated from the internalized membrane and synaptic endosomes. Furthermore, these diverse modes of endocytosis appear to influence both the molecular composition and subsequent physiological role of individual SVs. In addition, previously unknown complexity in SV refilling and reclustering has been revealed. This review presents a modern view of the SV life cycle and discusses how neuronal subtype, physiological temperature, and individual activity patterns can recruit different endocytic modes to generate new SVs and sculpt subsequent presynaptic performance.
Collapse
|
19
|
Endocytic Adaptor Proteins in Health and Disease: Lessons from Model Organisms and Human Mutations. Cells 2019; 8:cells8111345. [PMID: 31671891 PMCID: PMC6912373 DOI: 10.3390/cells8111345] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
Cells need to exchange material and information with their environment. This is largely achieved via cell-surface receptors which mediate processes ranging from nutrient uptake to signaling responses. Consequently, their surface levels have to be dynamically controlled. Endocytosis constitutes a powerful mechanism to regulate the surface proteome and to recycle vesicular transmembrane proteins that strand at the plasma membrane after exocytosis. For efficient internalization, the cargo proteins need to be linked to the endocytic machinery via adaptor proteins such as the heterotetrameric endocytic adaptor complex AP-2 and a variety of mostly monomeric endocytic adaptors. In line with the importance of endocytosis for nutrient uptake, cell signaling and neurotransmission, animal models and human mutations have revealed that defects in these adaptors are associated with several diseases ranging from metabolic disorders to encephalopathies. This review will discuss the physiological functions of the so far known adaptor proteins and will provide a comprehensive overview of their links to human diseases.
Collapse
|
20
|
Kokotos AC, Harper CB, Marland JRK, Smillie KJ, Cousin MA, Gordon SL. Synaptophysin sustains presynaptic performance by preserving vesicular synaptobrevin-II levels. J Neurochem 2019; 151:28-37. [PMID: 31216055 PMCID: PMC6851701 DOI: 10.1111/jnc.14797] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/24/2019] [Accepted: 06/01/2019] [Indexed: 01/01/2023]
Abstract
The two most abundant molecules on synaptic vesicles (SVs) are synaptophysin and synaptobrevin‐II (sybII). SybII is essential for SV fusion, whereas synaptophysin is proposed to control the trafficking of sybII after SV fusion and its retrieval during endocytosis. Despite controlling key aspects of sybII packaging into SVs, the absence of synaptophysin results in negligible effects on neurotransmission. We hypothesised that this apparent absence of effect may be because of the abundance of sybII on SVs, with the impact of inefficient sybII retrieval only revealed during periods of repeated SV turnover. To test this hypothesis, we subjected primary cultures of synaptophysin knockout neurons to repeated trains of neuronal activity, while monitoring SV fusion events and levels of vesicular sybII. We identified a significant decrease in both the number of SV fusion events (monitored using the genetically encoded reporter vesicular glutamate transporter‐pHluorin) and vesicular sybII levels (via both immunofluorescence and Western blotting) using this protocol. This revealed that synaptophysin is essential to sustain both parameters during periods of repetitive SV turnover. This was confirmed by the rescue of presynaptic performance by the expression of exogenous synaptophysin. Importantly, the expression of exogenous sybII also fully restored SV fusion events in synaptophysin knockout neurons. The ability of additional copies of sybII to fully rescue presynaptic performance in these knockout neurons suggests that the principal role of synaptophysin is to mediate the efficient retrieval of sybII to sustain neurotransmitter release. ![]()
Collapse
Affiliation(s)
- Alexandros C Kokotos
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh, Scotland, UK, EH8 9XD.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, UK, EH8 9XD
| | - Callista B Harper
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh, Scotland, UK, EH8 9XD.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, UK, EH8 9XD.,Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, UK, EH8 9XD
| | - Jamie R K Marland
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh, Scotland, UK, EH8 9XD
| | - Karen J Smillie
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh, Scotland, UK, EH8 9XD.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, UK, EH8 9XD
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, George Square, Edinburgh, Scotland, UK, EH8 9XD.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, UK, EH8 9XD.,Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, UK, EH8 9XD
| | - Sarah L Gordon
- The Florey Institute of Neuroscience and Mental Health, and Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Kroll J, Özçete ÖD, Jung S, Maritzen T, Milosevic I, Wichmann C, Moser T. AP180 promotes release site clearance and clathrin-dependent vesicle reformation in mouse cochlear inner hair cells. J Cell Sci 2019; 133:jcs.236737. [DOI: 10.1242/jcs.236737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/09/2019] [Indexed: 12/18/2022] Open
Abstract
High-throughput neurotransmission at ribbon synapses of cochlear inner hair cells (IHCs) requires tight coupling of neurotransmitter release and balanced recycling of synaptic vesicles (SVs) as well as rapid restoration of release sites. Here, we examined the role of the adaptor protein AP180 for IHC synaptic transmission in AP180-KO mice using high-pressure freezing and electron tomography, confocal microscopy, patch-clamp membrane-capacitance measurements and systems physiology. AP180 was found predominantly at the synaptic pole of IHCs. AP180-deficient IHCs had severely reduced SV numbers, slowed endocytic membrane retrieval, and accumulated endocytic intermediates near ribbon synapses, indicating that AP180 is required for clathrin-dependent endocytosis and SV reformation in IHCs. Moreover, AP180 deletion led to a high prevalence of SVs in a multi-tethered or docked state after stimulation, a reduced rate of SV replenishment, and a hearing impairment. We conclude that, in addition to its role in clathrin recruitment, AP180 contributes to release site clearance in IHCs.
Collapse
Affiliation(s)
- Jana Kroll
- Synaptic Vesicle Dynamics Group, European Neuroscience Institute Göttingen – A Joint Initiative of the University Medical Center Göttingen and the Max-Planck-Society, Göttingen, Germany
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Özge Demet Özçete
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sangyong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Neuro Modulation and Neuro Circuitry Group, Singapore Bioimaging Consortium (SBIC), Biomedical Sciences Institutes, 138667 Singapore
| | - Tanja Maritzen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Ira Milosevic
- Synaptic Vesicle Dynamics Group, European Neuroscience Institute Göttingen – A Joint Initiative of the University Medical Center Göttingen and the Max-Planck-Society, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Moser
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
22
|
Julien C, Tomberlin C, Roberts CM, Akram A, Stein GH, Silverman MA, Link CD. In vivo induction of membrane damage by β-amyloid peptide oligomers. Acta Neuropathol Commun 2018; 6:131. [PMID: 30497524 PMCID: PMC6263551 DOI: 10.1186/s40478-018-0634-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/13/2018] [Indexed: 01/17/2023] Open
Abstract
Exposure to the β-amyloid peptide (Aβ) is toxic to neurons and other cell types, but the mechanism(s) involved are still unresolved. Synthetic Aβ oligomers can induce ion-permeable pores in synthetic membranes, but whether this ability to damage membranes plays a role in the ability of Aβ oligomers to induce tau hyperphosphorylation, or other disease-relevant pathological changes, is unclear. To examine the cellular responses to Aβ exposure independent of possible receptor interactions, we have developed an in vivo C. elegans model that allows us to visualize these cellular responses in living animals. We find that feeding C. elegans E. coli expressing human Aβ induces a membrane repair response similar to that induced by exposure to the CRY5B, a known pore-forming toxin produced by B. thuringensis. This repair response does not occur when C. elegans is exposed to an Aβ Gly37Leu variant, which we have previously shown to be incapable of inducing tau phosphorylation in hippocampal neurons. The repair response is also blocked by loss of calpain function, and is altered by loss-of-function mutations in the C. elegans orthologs of BIN1 and PICALM, well-established risk genes for late onset Alzheimer's disease. To investigate the role of membrane repair on tau phosphorylation directly, we exposed hippocampal neurons to streptolysin O (SLO), a pore-forming toxin that induces a well-characterized membrane repair response. We find that SLO induces tau hyperphosphorylation, which is blocked by calpain inhibition. Finally, we use a novel biarsenical dye-tagging approach to show that the Gly37Leu substitution interferes with Aβ multimerization and thus the formation of potentially pore-forming oligomers. We propose that Aβ-induced tau hyperphosphorylation may be a downstream consequence of induction of a membrane repair process.
Collapse
|
23
|
Blazie SM, Jin Y. Pharming for Genes in Neurotransmission: Combining Chemical and Genetic Approaches in Caenorhabditis elegans. ACS Chem Neurosci 2018; 9:1963-1974. [PMID: 29432681 DOI: 10.1021/acschemneuro.7b00509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Synaptic transmission is central to nervous system function. Chemical and genetic screens are valuable approaches to probe synaptic mechanisms in living animals. The nematode Caenorhabditis elegans is a prime system to apply these methods to discover genes and dissect the cellular pathways underlying neurotransmission. Here, we review key approaches to understand neurotransmission and the action of psychiatric drugs in C. elegans. We start with early studies on cholinergic excitatory signaling at the neuromuscular junction, and move into mechanisms mediated by biogenic amines. Finally, we discuss emerging work toward understanding the mechanisms driving synaptic plasticity with a focus on regulation of protein translation.
Collapse
Affiliation(s)
- Stephen M. Blazie
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Yishi Jin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
24
|
Gan Q, Watanabe S. Synaptic Vesicle Endocytosis in Different Model Systems. Front Cell Neurosci 2018; 12:171. [PMID: 30002619 PMCID: PMC6031744 DOI: 10.3389/fncel.2018.00171] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/01/2018] [Indexed: 11/13/2022] Open
Abstract
Neurotransmission in complex animals depends on a choir of functionally distinct synapses releasing neurotransmitters in a highly coordinated manner. During synaptic signaling, vesicles fuse with the plasma membrane to release their contents. The rate of vesicle fusion is high and can exceed the rate at which synaptic vesicles can be re-supplied by distant sources. Thus, local compensatory endocytosis is needed to replenish the synaptic vesicle pools. Over the last four decades, various experimental methods and model systems have been used to study the cellular and molecular mechanisms underlying synaptic vesicle cycle. Clathrin-mediated endocytosis is thought to be the predominant mechanism for synaptic vesicle recycling. However, recent studies suggest significant contribution from other modes of endocytosis, including fast compensatory endocytosis, activity-dependent bulk endocytosis, ultrafast endocytosis, as well as kiss-and-run. Currently, it is not clear whether a universal model of vesicle recycling exist for all types of synapses. It is possible that each synapse type employs a particular mode of endocytosis. Alternatively, multiple modes of endocytosis operate at the same synapse, and the synapse toggles between different modes depending on its activity level. Here we compile review and research articles based on well-characterized model systems: frog neuromuscular junctions, C. elegans neuromuscular junctions, Drosophila neuromuscular junctions, lamprey reticulospinal giant axons, goldfish retinal ribbon synapses, the calyx of Held, and rodent hippocampal synapses. We will compare these systems in terms of their known modes and kinetics of synaptic vesicle endocytosis, as well as the underlying molecular machineries. We will also provide the future development of this field.
Collapse
Affiliation(s)
- Quan Gan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
25
|
Takatori S, Tomita T. AP180 N-Terminal Homology (ANTH) and Epsin N-Terminal Homology (ENTH) Domains: Physiological Functions and Involvement in Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:55-76. [DOI: 10.1007/5584_2018_218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
26
|
Ventimiglia D, Bargmann CI. Diverse modes of synaptic signaling, regulation, and plasticity distinguish two classes of C. elegans glutamatergic neurons. eLife 2017; 6:e31234. [PMID: 29160768 PMCID: PMC5705214 DOI: 10.7554/elife.31234] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/20/2017] [Indexed: 11/13/2022] Open
Abstract
Synaptic vesicle release properties vary between neuronal cell types, but in most cases the molecular basis of this heterogeneity is unknown. Here, we compare in vivo synaptic properties of two neuronal classes in the C. elegans central nervous system, using VGLUT-pHluorin to monitor synaptic vesicle exocytosis and retrieval in intact animals. We show that the glutamatergic sensory neurons AWCON and ASH have distinct synaptic dynamics associated with tonic and phasic synaptic properties, respectively. Exocytosis in ASH and AWCON is differentially affected by SNARE-complex regulators that are present in both neurons: phasic ASH release is strongly dependent on UNC-13, whereas tonic AWCON release relies upon UNC-18 and on the protein kinase C homolog PKC-1. Strong stimuli that elicit high calcium levels increase exocytosis and retrieval rates in AWCON, generating distinct tonic and evoked synaptic modes. These results highlight the differential deployment of shared presynaptic proteins in neuronal cell type-specific functions.
Collapse
Affiliation(s)
- Donovan Ventimiglia
- Lulu and Anthony Wang Laboratory of Neural Circuits and BehaviorThe Rockefeller UniversityNew YorkUnited States
| | - Cornelia I Bargmann
- Lulu and Anthony Wang Laboratory of Neural Circuits and BehaviorThe Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
27
|
Kaempf N, Maritzen T. Safeguards of Neurotransmission: Endocytic Adaptors as Regulators of Synaptic Vesicle Composition and Function. Front Cell Neurosci 2017; 11:320. [PMID: 29085282 PMCID: PMC5649181 DOI: 10.3389/fncel.2017.00320] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Communication between neurons relies on neurotransmitters which are released from synaptic vesicles (SVs) upon Ca2+ stimuli. To efficiently load neurotransmitters, sense the rise in intracellular Ca2+ and fuse with the presynaptic membrane, SVs need to be equipped with a stringently controlled set of transmembrane proteins. In fact, changes in SV protein composition quickly compromise neurotransmission and most prominently give rise to epileptic seizures. During exocytosis SVs fully collapse into the presynaptic membrane and consequently have to be replenished to sustain neurotransmission. Therefore, surface-stranded SV proteins have to be efficiently retrieved post-fusion to be used for the generation of a new set of fully functional SVs, a process in which dedicated endocytic sorting adaptors play a crucial role. The question of how the precise reformation of SVs is achieved is intimately linked to how SV membranes are retrieved. For a long time both processes were believed to be two sides of the same coin since Clathrin-mediated endocytosis (CME), the proposed predominant SV recycling mode, will jointly retrieve SV membranes and proteins. However, with the recent proposal of Clathrin-independent SV recycling pathways SV membrane retrieval and SV reformation turn into separable events. This review highlights the progress made in unraveling the molecular mechanisms mediating the high-fidelity retrieval of SV proteins and discusses how the gathered knowledge about SV protein recycling fits in with the new notions of SV membrane endocytosis.
Collapse
Affiliation(s)
- Natalie Kaempf
- Molecular Physiology and Cell Biology Section, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Tanja Maritzen
- Molecular Physiology and Cell Biology Section, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| |
Collapse
|
28
|
Tyson T, Senchuk M, Cooper JF, George S, Van Raamsdonk JM, Brundin P. Novel animal model defines genetic contributions for neuron-to-neuron transfer of α-synuclein. Sci Rep 2017; 7:7506. [PMID: 28790319 PMCID: PMC5548897 DOI: 10.1038/s41598-017-07383-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 06/22/2017] [Indexed: 12/27/2022] Open
Abstract
Cell-to-cell spreading of misfolded α-synuclein (α-syn) is suggested to contribute to the progression of neuropathology in Parkinson’s disease (PD). Compelling evidence supports the hypothesis that misfolded α-syn transmits from neuron-to-neuron and seeds aggregation of the protein in the recipient cells. Furthermore, α-syn frequently appears to propagate in the brains of PD patients following a stereotypic pattern consistent with progressive spreading along anatomical pathways. We have generated a C. elegans model that mirrors this progression and allows us to monitor α-syn neuron-to-neuron transmission in a live animal over its lifespan. We found that modulation of autophagy or exo/endocytosis, affects α-syn transfer. Furthermore, we demonstrate that silencing C. elegans orthologs of PD-related genes also increases the accumulation of α-syn. This novel worm model is ideal for screening molecules and genes to identify those that modulate prion-like spreading of α-syn in order to target novel strategies for disease modification in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Trevor Tyson
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA.
| | - Megan Senchuk
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jason F Cooper
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Sonia George
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jeremy M Van Raamsdonk
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
29
|
Han M, Zou W, Chang H, Yu Y, Zhang H, Li S, Cheng H, Wei G, Chen Y, Reinke V, Xu T, Kang L. A Systematic RNAi Screen Reveals a Novel Role of a Spindle Assembly Checkpoint Protein BuGZ in Synaptic Transmission in C. elegans. Front Mol Neurosci 2017; 10:141. [PMID: 28553202 PMCID: PMC5425591 DOI: 10.3389/fnmol.2017.00141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/25/2017] [Indexed: 11/29/2022] Open
Abstract
Synaptic vesicles (SV) store various neurotransmitters that are released at the synapse. The molecular mechanisms of biogenesis, exocytosis, and endocytosis for SV, however, remain largely elusive. In this study, using Complex Object Parametric Analysis and Sorter (COPAS) to monitor the fluorescence of synapto-pHluorin (SpH), we performed a whole-genome RNAi screen in C. elegans to identify novel genetic modulators in SV cycling. One hundred seventy six genes that up-regulating SpH fluorescence and 96 genes that down-regulating SpH fluorescence were identified after multi-round screen. Among these genes, B0035.1 (bugz-1) encodes ortholog of mammalian C2H2 zinc-finger protein BuGZ/ZNF207, which is a spindle assembly checkpoint protein essential for mitosis in human cells. Combining electrophysiology, imaging and behavioral assays, we reveal that depletion of BuGZ-1 results in defects in locomotion. We further demonstrate that BuGZ-1 promotes SV recycling by regulating the expression levels of endocytosis-related genes such as rab11.1. Therefore, we have identified a bunch of potential genetic modulators in SV cycling, and revealed an unexpected role of BuGZ-1 in regulating synaptic transmission.
Collapse
Affiliation(s)
- Mei Han
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Department of Neurobiology, Institute of Neuroscience, Zhejiang University School of MedicineHangzhou, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China.,Department of Genetics, Yale University School of MedicineNew Haven, CT, USA
| | - Wenjuan Zou
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Department of Neurobiology, Institute of Neuroscience, Zhejiang University School of MedicineHangzhou, China
| | - Hao Chang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China.,Department of Genetics, Yale University School of MedicineNew Haven, CT, USA
| | - Yong Yu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
| | - Haining Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
| | - Shitian Li
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Department of Neurobiology, Institute of Neuroscience, Zhejiang University School of MedicineHangzhou, China
| | - Hankui Cheng
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Department of Neurobiology, Institute of Neuroscience, Zhejiang University School of MedicineHangzhou, China
| | - Guifeng Wei
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
| | - Yan Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
| | - Valerie Reinke
- Department of Genetics, Yale University School of MedicineNew Haven, CT, USA
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
| | - Lijun Kang
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Department of Neurobiology, Institute of Neuroscience, Zhejiang University School of MedicineHangzhou, China
| |
Collapse
|
30
|
Steuer Costa W, Yu SC, Liewald JF, Gottschalk A. Fast cAMP Modulation of Neurotransmission via Neuropeptide Signals and Vesicle Loading. Curr Biol 2017; 27:495-507. [PMID: 28162892 DOI: 10.1016/j.cub.2016.12.055] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 12/06/2016] [Accepted: 12/27/2016] [Indexed: 02/02/2023]
Abstract
Cyclic AMP (cAMP) signaling augments synaptic transmission, but because many targets of cAMP and protein kinase A (PKA) may be involved, mechanisms underlying this pathway remain unclear. To probe this mechanism, we used optogenetic stimulation of cAMP signaling by Beggiatoa-photoactivated adenylyl cyclase (bPAC) in Caenorhabditis elegans motor neurons. Behavioral, electron microscopy (EM), and electrophysiology analyses revealed cAMP effects on both the rate and on quantal size of transmitter release and led to the identification of a neuropeptidergic pathway affecting quantal size. cAMP enhanced synaptic vesicle (SV) fusion by increasing mobilization and docking/priming. cAMP further evoked dense core vesicle (DCV) release of neuropeptides, in contrast to channelrhodopsin (ChR2) stimulation. cAMP-evoked DCV release required UNC-31/Ca2+-dependent activator protein for secretion (CAPS). Thus, DCVs accumulated in unc-31 mutant synapses. bPAC-induced neuropeptide signaling acts presynaptically to enhance vAChT-dependent SV loading with acetylcholine, thus causing increased miniature postsynaptic current amplitudes (mPSCs) and significantly enlarged SVs.
Collapse
Affiliation(s)
- Wagner Steuer Costa
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, 60438 Frankfurt, Germany; Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Szi-Chieh Yu
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, 60438 Frankfurt, Germany; Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Jana F Liewald
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, 60438 Frankfurt, Germany; Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, 60438 Frankfurt, Germany; Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany.
| |
Collapse
|
31
|
Pucadyil TJ, Holkar SS. Comparative analysis of adaptor-mediated clathrin assembly reveals general principles for adaptor clustering. Mol Biol Cell 2016; 27:3156-3163. [PMID: 27559129 PMCID: PMC5063622 DOI: 10.1091/mbc.e16-06-0399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/19/2016] [Indexed: 11/30/2022] Open
Abstract
Clathrin-mediated endocytosis sorts the bulk of membrane proteins and is a process that starts with adaptor-induced clathrin assembly. Real-time fluorescence analysis shows that adaptor sorting is determined not by the extent of clathrin recruited or the degree of clathrin clustered but instead by the rate of clathrin assembly. Clathrin-mediated endocytosis (CME) manages the sorting and uptake of the bulk of membrane proteins (or cargo) from the plasma membrane. CME is initiated by the formation of clathrin-coated pits (CCPs), in which adaptors nucleate clathrin assembly. Clathrin adaptors display diversity in both the type and number of evolutionarily conserved clathrin-binding boxes. How this diversity relates to the process of adaptor clustering as clathrin assembles around a growing pit remains unclear. Using real-time, fluorescence microscopy–based assays, we compare the formation kinetics and distribution of clathrin assemblies on membranes that display five unique clathrin adaptors. Correlations between equilibrium and kinetic parameters of clathrin assembly to the eventual adaptor distribution indicate that adaptor clustering is determined not by the amount of clathrin recruited or the degree of clathrin clustered but instead by the rate of clathrin assembly. Together our results emphasize the need to analyze kinetics of protein interactions to better understand mechanisms that regulate CME.
Collapse
Affiliation(s)
- Thomas J Pucadyil
- Indian Institute of Science Education and Research, Pune, Maharashtra 411 008, India
| | - Sachin S Holkar
- Indian Institute of Science Education and Research, Pune, Maharashtra 411 008, India
| |
Collapse
|
32
|
Kobayashi S, Hida Y, Ishizaki H, Inoue E, Tanaka-Okamoto M, Yamasaki M, Miyazaki T, Fukaya M, Kitajima I, Takai Y, Watanabe M, Ohtsuka T, Manabe T. The active zone protein CAST regulates synaptic vesicle recycling and quantal size in the mouse hippocampus. Eur J Neurosci 2016; 44:2272-84. [PMID: 27422015 DOI: 10.1111/ejn.13331] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 01/13/2023]
Abstract
Synaptic efficacy is determined by various factors, including the quantal size, which is dependent on the amount of neurotransmitters in synaptic vesicles at the presynaptic terminal. It is essential for stable synaptic transmission that the quantal size is kept within a constant range and that synaptic efficacy during and after repetitive synaptic activation is maintained by replenishing release sites with synaptic vesicles. However, the mechanisms for these fundamental properties have still been undetermined. We found that the active zone protein CAST (cytomatrix at the active zone structural protein) played pivotal roles in both presynaptic regulation of quantal size and recycling of endocytosed synaptic vesicles. In the CA1 region of hippocampal slices of the CAST knockout mice, miniature excitatory synaptic responses were increased in size, and synaptic depression after prolonged synaptic activation was larger, which was attributable to selective impairment of synaptic vesicle trafficking via the endosome in the presynaptic terminal likely mediated by Rab6. Therefore, CAST serves as a key molecule that regulates dynamics and neurotransmitter contents of synaptic vesicles in the excitatory presynaptic terminal in the central nervous system.
Collapse
Affiliation(s)
- Shizuka Kobayashi
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Yamato Hida
- Department of Biochemistry, University of Yamanashi, Chuo, 409-3898, Japan
| | | | | | - Miki Tanaka-Okamoto
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Miwako Yamasaki
- Department of Anatomy and Embryology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Taisuke Miyazaki
- Department of Anatomy and Embryology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiro Fukaya
- Department of Anatomy and Embryology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Isao Kitajima
- Department of Clinical Laboratory and Molecular Pathology, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Yoshimi Takai
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiko Watanabe
- Department of Anatomy and Embryology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, University of Yamanashi, Chuo, 409-3898, Japan
| | - Toshiya Manabe
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| |
Collapse
|
33
|
The Conserved VPS-50 Protein Functions in Dense-Core Vesicle Maturation and Acidification and Controls Animal Behavior. Curr Biol 2016; 26:862-71. [PMID: 26948874 DOI: 10.1016/j.cub.2016.01.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 11/20/2015] [Accepted: 01/21/2016] [Indexed: 02/05/2023]
Abstract
The modification of behavior in response to experience is crucial for animals to adapt to environmental changes. Although factors such as neuropeptides and hormones are known to function in the switch between alternative behavioral states, the mechanisms by which these factors transduce, store, retrieve, and integrate environmental signals to regulate behavior are poorly understood. The rate of locomotion of the nematode Caenorhabditis elegans depends on both current and past food availability. Specifically, C. elegans slows its locomotion when it encounters food, and animals in a food-deprived state slow even more than animals in a well-fed state. The slowing responses of well-fed and food-deprived animals in the presence of food represent distinct behavioral states, as they are controlled by different sets of genes, neurotransmitters, and neurons. Here we describe an evolutionarily conserved C. elegans protein, VPS-50, that is required for animals to assume the well-fed behavioral state. Both VPS-50 and its murine homolog mVPS50 are expressed in neurons, are associated with synaptic and dense-core vesicles, and control vesicle acidification and hence synaptic function, likely through regulation of the assembly of the V-ATPase complex. We propose that dense-core vesicle acidification controlled by the evolutionarily conserved protein VPS-50/mVPS50 affects behavioral state by modulating neuropeptide levels and presynaptic neuronal function in both C. elegans and mammals.
Collapse
|
34
|
Li LB, Lei H, Arey RN, Li P, Liu J, Murphy CT, Xu XZS, Shen K. The Neuronal Kinesin UNC-104/KIF1A Is a Key Regulator of Synaptic Aging and Insulin Signaling-Regulated Memory. Curr Biol 2016; 26:605-15. [PMID: 26877087 DOI: 10.1016/j.cub.2015.12.068] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/23/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022]
Abstract
Aging is the greatest risk factor for a number of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease. Furthermore, normal aging is associated with a decline in sensory, motor, and cognitive functions. Emerging evidence suggests that synapse alterations, rather than neuronal cell death, are the causes of neuronal dysfunctions in normal aging and in early stages of neurodegenerative diseases. However, little is known about the mechanisms underlying age-related synaptic decline. Here, we uncover a surprising role of the anterograde molecular motor UNC-104/KIF1A as a key regulator of neural circuit deterioration in aging C. elegans. Through analyses of synapse protein localization, synaptic transmission, and animal behaviors, we find that reduced function of UNC-104 accelerates motor circuit dysfunction with age, whereas upregulation of UNC-104 significantly improves motor function at advanced ages and also mildly extends lifespan. In addition, UNC-104-overexpressing animals outperform wild-type controls in associative learning and memory tests. Further genetic analyses suggest that UNC-104 functions downstream of the DAF-2-signaling pathway and is regulated by the FOXO transcription factor DAF-16, which contributes to the effects of DAF-2 in neuronal aging. Together, our cellular, electrophysiological, and behavioral analyses highlight the importance of axonal transport in the maintenance of synaptic structural integrity and function during aging and raise the possibility of targeting kinesins to slow age-related neural circuit dysfunction.
Collapse
Affiliation(s)
- Ling-Bo Li
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Haoyun Lei
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; College of Life Science and Technology, Huazhong University of Science Technology, Wuhan, Hubei 430074, China
| | - Rachel N Arey
- Department of Molecular Biology and LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Pengpeng Li
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jianfeng Liu
- College of Life Science and Technology, Huazhong University of Science Technology, Wuhan, Hubei 430074, China
| | - Coleen T Murphy
- Department of Molecular Biology and LSI Genomics, Princeton University, Princeton, NJ 08544, USA.
| | - X Z Shawn Xu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Kang Shen
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
35
|
Gordon SL, Cousin MA. The iTRAPs: Guardians of Synaptic Vesicle Cargo Retrieval During Endocytosis. Front Synaptic Neurosci 2016; 8:1. [PMID: 26903854 PMCID: PMC4746236 DOI: 10.3389/fnsyn.2016.00001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/25/2016] [Indexed: 11/30/2022] Open
Abstract
The reformation of synaptic vesicles (SVs) during endocytosis is essential for the maintenance of neurotransmission in central nerve terminals. Newly formed SVs must be generated with the correct protein cargo in the correct stoichiometry to be functional for exocytosis. Classical clathrin adaptor protein complexes play a key role in sorting and clustering synaptic vesicle cargo in this regard. However it is becoming increasingly apparent that additional “fail-safe” mechanisms exist to ensure the accurate retrieval of essential cargo molecules. For example, the monomeric adaptor proteins AP180/CALM and stonin-2 are required for the efficient retrieval of synaptobrevin II (sybII) and synaptotagmin-1 respectively. Furthermore, recent studies have revealed that sybII and synaptotagmin-1 interact with other SV cargoes to ensure a high fidelity of retrieval. These cargoes are synaptophysin (for sybII) and SV2A (for synaptotagmin-1). In this review, we summarize current knowledge regarding the retrieval mechanisms for both sybII and synaptotagmin-1 during endocytosis. We also define and set criteria for a new functional group of SV molecules that facilitate the retrieval of their interaction partners. We have termed these molecules intrinsic trafficking partners (iTRAPs) and we discuss how the function of this group impacts on presynaptic performance in both health and disease.
Collapse
Affiliation(s)
- Sarah L Gordon
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Michael A Cousin
- Centre for Integrative Physiology, University of Edinburgh Edinburgh, UK
| |
Collapse
|
36
|
de Aragão BC, Rodrigues HA, Valadão PAC, Camargo W, Naves LA, Ribeiro FM, Guatimosim C. Changes in structure and function of diaphragm neuromuscular junctions from BACHD mouse model for Huntington's disease. Neurochem Int 2016; 93:64-72. [DOI: 10.1016/j.neuint.2015.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/17/2015] [Accepted: 12/28/2015] [Indexed: 10/22/2022]
|
37
|
Robinson MS. Forty Years of Clathrin-coated Vesicles. Traffic 2015; 16:1210-38. [PMID: 26403691 DOI: 10.1111/tra.12335] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
Abstract
The purification of coated vesicles and the discovery of clathrin by Barbara Pearse in 1975 was a landmark in cell biology. Over the past 40 years, work from many labs has uncovered the molecular details of clathrin and its associated proteins, including how they assemble into a coated vesicle and how they select cargo. Unexpected connections have been found with signalling, development, neuronal transmission, infection, immunity and genetic disorders. But there are still a number of unanswered questions, including how clathrin-mediated trafficking is regulated and how the machinery evolved.
Collapse
Affiliation(s)
- Margaret S Robinson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
38
|
Koo SJ, Kochlamazashvili G, Rost B, Puchkov D, Gimber N, Lehmann M, Tadeus G, Schmoranzer J, Rosenmund C, Haucke V, Maritzen T. Vesicular Synaptobrevin/VAMP2 Levels Guarded by AP180 Control Efficient Neurotransmission. Neuron 2015; 88:330-44. [PMID: 26412491 DOI: 10.1016/j.neuron.2015.08.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 07/17/2015] [Accepted: 08/19/2015] [Indexed: 01/01/2023]
Abstract
Neurotransmission depends on synaptic vesicle (SV) exocytosis driven by soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex formation of vesicular synaptobrevin/VAMP2 (Syb2). Exocytic fusion is followed by endocytic SV membrane retrieval and the high-fidelity reformation of SVs. Syb2 is the most abundant SV protein with 70 copies per SV, yet, one to three Syb2 molecules appear to be sufficient for basal exocytosis. Here we demonstrate that loss of the Syb2-specific endocytic adaptor AP180 causes a moderate activity-dependent reduction of vesicular Syb2 levels, defects in SV reformation, and a corresponding impairment of neurotransmission that lead to excitatory/inhibitory imbalance, epileptic seizures, and premature death. Further reduction of Syb2 levels in AP180(-/-)/Syb2(+/-) mice results in perinatal lethality, whereas Syb2(+/-) mice partially phenocopy loss of AP180, indicating that reduced vesicular Syb2 levels underlie the observed defects in neurotransmission. Thus, a large vesicular Syb2 pool maintained by AP180 is crucial to sustain efficient neurotransmission and SV reformation.
Collapse
Affiliation(s)
- Seong Joo Koo
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany
| | - Gaga Kochlamazashvili
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany
| | - Benjamin Rost
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Dmytro Puchkov
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany
| | - Niclas Gimber
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany
| | - Martin Lehmann
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany; Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195 Berlin, Germany
| | - Georgi Tadeus
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany
| | - Jan Schmoranzer
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany; Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195 Berlin, Germany
| | - Christian Rosenmund
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Volker Haucke
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany; Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany.
| | - Tanja Maritzen
- Leibniz-Institut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
39
|
Miller SE, Mathiasen S, Bright NA, Pierre F, Kelly BT, Kladt N, Schauss A, Merrifield CJ, Stamou D, Höning S, Owen DJ. CALM regulates clathrin-coated vesicle size and maturation by directly sensing and driving membrane curvature. Dev Cell 2015; 33:163-75. [PMID: 25898166 PMCID: PMC4406947 DOI: 10.1016/j.devcel.2015.03.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 01/23/2015] [Accepted: 03/01/2015] [Indexed: 02/06/2023]
Abstract
The size of endocytic clathrin-coated vesicles (CCVs) is remarkably uniform, suggesting that it is optimized to achieve the appropriate levels of cargo and lipid internalization. The three most abundant proteins in mammalian endocytic CCVs are clathrin and the two cargo-selecting, clathrin adaptors, CALM and AP2. Here we demonstrate that depletion of CALM causes a substantial increase in the ratio of “open” clathrin-coated pits (CCPs) to “necked”/“closed” CCVs and a doubling of CCP/CCV diameter, whereas AP2 depletion has opposite effects. Depletion of either adaptor, however, significantly inhibits endocytosis of transferrin and epidermal growth factor. The phenotypic effects of CALM depletion can be rescued by re-expression of wild-type CALM, but not with CALM that lacks a functional N-terminal, membrane-inserting, curvature-sensing/driving amphipathic helix, the existence and properties of which are demonstrated. CALM is thus a major factor in controlling CCV size and maturation and hence in determining the rates of endocytic cargo uptake. CALM loss increases size and frequency of early endocytic clathrin-coated structures Depletion of CALM slows endocytic clathrin-coated pit maturation and endocytic rate CALM possesses an N-terminal, membrane-curvature-sensing/driving amphipathic helix Clathrin-coated pit maturation is regulated by CALM’s N-terminal amphipathic helix
Collapse
Affiliation(s)
- Sharon E Miller
- Cambridge Institute for Medical Research and Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK.
| | - Signe Mathiasen
- Bionanotechnology and Nanomedicine Laboratory, Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Nicholas A Bright
- Cambridge Institute for Medical Research and Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - Fabienne Pierre
- Laboratoire d'Enzymologie et Biochimie Structurales, UPR3082 CNRS - Bat 34, Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Bernard T Kelly
- Cambridge Institute for Medical Research and Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - Nikolay Kladt
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Astrid Schauss
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Christien J Merrifield
- Laboratoire d'Enzymologie et Biochimie Structurales, UPR3082 CNRS - Bat 34, Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Dimitrios Stamou
- Bionanotechnology and Nanomedicine Laboratory, Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Stefan Höning
- Institute of Biochemistry I and Center for Molecular Medicine Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany
| | - David J Owen
- Cambridge Institute for Medical Research and Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
40
|
Busch DJ, Houser JR, Hayden CC, Sherman MB, Lafer EM, Stachowiak JC. Intrinsically disordered proteins drive membrane curvature. Nat Commun 2015. [PMID: 26204806 PMCID: PMC4515776 DOI: 10.1038/ncomms8875] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Assembly of highly curved membrane structures is essential to cellular physiology. The prevailing view has been that proteins with curvature-promoting structural motifs, such as wedge-like amphipathic helices and crescent-shaped BAR domains, are required for bending membranes. Here we report that intrinsically disordered domains of the endocytic adaptor proteins, Epsin1 and AP180 are highly potent drivers of membrane curvature. This result is unexpected since intrinsically disordered domains lack a well-defined three-dimensional structure. However, in vitro measurements of membrane curvature and protein diffusivity demonstrate that the large hydrodynamic radii of these domains generate steric pressure that drives membrane bending. When disordered adaptor domains are expressed as transmembrane cargo in mammalian cells, they are excluded from clathrin-coated pits. We propose that a balance of steric pressure on the two surfaces of the membrane drives this exclusion. These results provide quantitative evidence for the influence of steric pressure on the content and assembly of curved cellular membrane structures. Proteins that bend membranes often contain curvature-promoting structural motifs such as wedges or crescent-shaped domains. Busch et al. report that intrinsically disordered domains can also drive membrane curvature and provide evidence that steric pressure driven by protein crowding mediates this effect.
Collapse
Affiliation(s)
- David J Busch
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, Texas 78712, USA
| | - Justin R Houser
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, Texas 78712, USA
| | - Carl C Hayden
- 1] Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, Texas 78712, USA [2] Sandia National Laboratories, 7011 East Avenue, Livermore, California 94550, USA
| | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 1.224 Medical Research Building, Galveston, Texas 77555, USA
| | - Eileen M Lafer
- Department of Biochemistry and Center for Biomedical Neuroscience, The University of Texas Health Science Center at San Antonio, UTHSCSA Biochemistry 415B, 7703 Floyd Curl Drive, San Antonio, Texas 78229, USA
| | - Jeanne C Stachowiak
- 1] Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, Texas 78712, USA [2] Institute for Cellular and Molecular Biology, The University of Texas at Austin, 107 W Dean, Keeton,Texas 78712, USA
| |
Collapse
|
41
|
Zhu M, Wu G, Li YX, Stevens JK, Fan CX, Spang A, Dong MQ. Serum- and Glucocorticoid-Inducible Kinase-1 (SGK-1) Plays a Role in Membrane Trafficking in Caenorhabditis elegans. PLoS One 2015; 10:e0130778. [PMID: 26115433 PMCID: PMC4482599 DOI: 10.1371/journal.pone.0130778] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/22/2015] [Indexed: 02/03/2023] Open
Abstract
The mammalian serum- and glucocorticoid-inducible kinase SGK1 regulates the endocytosis of ion channels. Here we report that in C. elegans sgk-1 null mutants, GFP-tagged MIG-14/Wntless, the sorting receptor of Wnt, failed to localize to the basolateral membrane of intestinal cells; instead, it was mis-sorted to lysosomes. This effect can be explained in part by altered sphingolipid levels, because reducing glucosylceramide biosynthesis restored the localization of MIG-14::GFP. Membrane traffic was not perturbed in general, as no obvious morphological defects were detected for early endosomes, the Golgi apparatus, and the endoplasmic reticulum (ER) in sgk-1 null animals. The recycling of MIG-14/Wntless through the Golgi might be partially responsible for the observed phenotype because the subcellular distribution of two plasma membrane cargoes that do not recycle through the trans-Golgi network (TGN) was affected to a lesser degree. Consistently, knockdown of the ArfGEF gbf-1 altered the distribution of SGK-1 at the basolateral membrane of intestinal cells. In addition, we found that sgk-1(RNAi) induced unfolded protein response in the ER, suggesting at least an indirect role of SGK-1 early in the secretory pathway. We propose that SGK-1 function is required for lipid homeostasis and that it acts at different intracellular trafficking steps.
Collapse
Affiliation(s)
- Ming Zhu
- College of Life Sciences, Beijing Normal University, Beijing, China
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Gang Wu
- National Institute of Biological Sciences, Beijing, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, National Institute of Biological Sciences, Beijing, Beijing, China
| | - Yu-Xin Li
- National Institute of Biological Sciences, Beijing, Beijing, China
| | | | - Chao-Xuan Fan
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Anne Spang
- Growth and Development, Biozentrum, University of Basel, Basel, Switzerland
| | - Meng-Qiu Dong
- College of Life Sciences, Beijing Normal University, Beijing, China
- National Institute of Biological Sciences, Beijing, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, National Institute of Biological Sciences, Beijing, Beijing, China
| |
Collapse
|
42
|
Kempf N, Didier P, Postupalenko V, Bucher B, Mély Y. Internalization mechanism of neuropeptide Y bound to its Y1receptor investigated by high resolution microscopy. Methods Appl Fluoresc 2015; 3:025004. [DOI: 10.1088/2050-6120/3/2/025004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
43
|
Garafalo SD, Luth ES, Moss BJ, Monteiro MI, Malkin E, Juo P. The AP2 clathrin adaptor protein complex regulates the abundance of GLR-1 glutamate receptors in the ventral nerve cord of Caenorhabditis elegans. Mol Biol Cell 2015; 26:1887-900. [PMID: 25788288 PMCID: PMC4436833 DOI: 10.1091/mbc.e14-06-1048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 03/06/2015] [Indexed: 01/23/2023] Open
Abstract
Regulation of glutamate receptor trafficking controls synaptic strength and plasticity. This study takes advantage of viable, null mutations in subunits of the clathrin adaptor protein 2 (AP2) complex in Caenorhabditis elegans to reveal a novel and unexpected AP2-dependent trafficking step for glutamate receptors early in the secretory pathway. Regulation of glutamate receptor (GluR) abundance at synapses by clathrin-mediated endocytosis can control synaptic strength and plasticity. We take advantage of viable, null mutations in subunits of the clathrin adaptor protein 2 (AP2) complex in Caenorhabditis elegans to characterize the in vivo role of AP2 in GluR trafficking. In contrast to our predictions for an endocytic adaptor, we found that levels of the GluR GLR-1 are decreased at synapses in the ventral nerve cord (VNC) of animals with mutations in the AP2 subunits APM-2/μ2, APA-2/α, or APS-2/σ2. Rescue experiments indicate that APM-2/μ2 functions in glr-1–expressing interneurons and the mature nervous system to promote GLR-1 levels in the VNC. Genetic analyses suggest that APM-2/μ2 acts upstream of GLR-1 endocytosis in the VNC. Consistent with this, GLR-1 accumulates in cell bodies of apm-2 mutants. However, GLR-1 does not appear to accumulate at the plasma membrane of the cell body as expected, but instead accumulates in intracellular compartments including Syntaxin-13– and RAB-14–labeled endosomes. This study reveals a novel role for the AP2 clathrin adaptor in promoting the abundance of GluRs at synapses in vivo, and implicates AP2 in the regulation of GluR trafficking at an early step in the secretory pathway.
Collapse
Affiliation(s)
- Steven D Garafalo
- Department of Developmental, Molecular & Chemical Biology Graduate Program in Cellular and Molecular Physiology, Tufts University School of Medicine, Boston, MA 02111
| | - Eric S Luth
- Department of Developmental, Molecular & Chemical Biology
| | - Benjamin J Moss
- Department of Developmental, Molecular & Chemical Biology Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Michael I Monteiro
- Department of Developmental, Molecular & Chemical Biology Graduate Program in Cellular and Molecular Physiology, Tufts University School of Medicine, Boston, MA 02111
| | - Emily Malkin
- Department of Developmental, Molecular & Chemical Biology
| | - Peter Juo
- Department of Developmental, Molecular & Chemical Biology
| |
Collapse
|
44
|
The ∼ 16 kDa C-terminal sequence of clathrin assembly protein AP180 is essential for efficient clathrin binding. PLoS One 2014; 9:e110557. [PMID: 25329427 PMCID: PMC4203807 DOI: 10.1371/journal.pone.0110557] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 09/22/2014] [Indexed: 11/19/2022] Open
Abstract
Brain-specific AP180 is present in clathrin coats at equal concentration to the adapter complex, AP2, and assembles clathrin faster than any other protein in vitro. Both AP180 and its ubiquitously expressed homolog clathrin assembly lymphoid myeloid leukemia protein (CALM) control vesicle size and shape in clathrin mediated endocytosis. The clathrin assembly role of AP180 is mediated by a long disordered C-terminal assembly domain. Within this assembly domain, a central acidic clathrin and adapter binding (CLAP) sub-domain contains all of the known short binding motifs for clathrin and AP2. The role of the remaining ∼ 16 kDa C-terminal sequence has not been clear. We show that this sequence has a separate function in ensuring efficient binding of clathrin, based on in vitro binding and ex vivo transferrin uptake assays. Sequence alignment suggests the C-terminal sub-domain is conserved in CALM.
Collapse
|
45
|
Watanabe S, Trimbuch T, Camacho-Pérez M, Rost BR, Brokowski B, Söhl-Kielczynski B, Felies A, Davis MW, Rosenmund C, Jorgensen EM. Clathrin regenerates synaptic vesicles from endosomes. Nature 2014; 515:228-33. [PMID: 25296249 PMCID: PMC4291189 DOI: 10.1038/nature13846] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022]
Abstract
Ultrafast endocytosis can retrieve a single, large endocytic vesicle as fast as 50-100 ms after synaptic vesicle fusion. However, the fate of the large endocytic vesicles is not known. Here we demonstrate that these vesicles transition to a synaptic endosome about one second after stimulation. The endosome is resolved into coated vesicles after 3 s, which in turn become small-diameter synaptic vesicles 5-6 s after stimulation. We disrupted clathrin function using RNA interference (RNAi) and found that clathrin is not required for ultrafast endocytosis but is required to generate synaptic vesicles from the endosome. Ultrafast endocytosis fails when actin polymerization is disrupted, or when neurons are stimulated at room temperature instead of physiological temperature. In the absence of ultrafast endocytosis, synaptic vesicles are retrieved directly from the plasma membrane by clathrin-mediated endocytosis. These results may explain discrepancies among published experiments concerning the role of clathrin in synaptic vesicle endocytosis.
Collapse
Affiliation(s)
- Shigeki Watanabe
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah 84112-0840, USA
| | - Thorsten Trimbuch
- Neuroscience Research Center Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Marcial Camacho-Pérez
- Neuroscience Research Center Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Benjamin R Rost
- 1] Neuroscience Research Center Charité Universitätsmedizin Berlin, Berlin 10117, Germany [2] German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Bettina Brokowski
- Neuroscience Research Center Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | | | - Annegret Felies
- Neuroscience Research Center Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - M Wayne Davis
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah 84112-0840, USA
| | - Christian Rosenmund
- Neuroscience Research Center Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Erik M Jorgensen
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah 84112-0840, USA
| |
Collapse
|
46
|
Moshkanbaryans L, Chan LS, Graham ME. The Biochemical Properties and Functions of CALM and AP180 in Clathrin Mediated Endocytosis. MEMBRANES 2014; 4:388-413. [PMID: 25090048 PMCID: PMC4194041 DOI: 10.3390/membranes4030388] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/03/2014] [Accepted: 07/22/2014] [Indexed: 01/26/2023]
Abstract
Clathrin-mediated endocytosis (CME) is a fundamental process for the regulated internalization of transmembrane cargo and ligands via the formation of vesicles using a clathrin coat. A vesicle coat is initially created at the plasma membrane by clathrin assembly into a lattice, while a specific cargo sorting process selects and concentrates proteins for inclusion in the new vesicle. Vesicles formed via CME traffic to different parts of the cell and fuse with target membranes to deliver cargo. Both clathrin assembly and cargo sorting functions are features of the two gene family consisting of assembly protein 180 kDa (AP180) and clathrin assembly lymphoid myeloid leukemia protein (CALM). In this review, we compare the primary structure and domain organization of CALM and AP180 and relate these properties to known functions and roles in CME and disease.
Collapse
Affiliation(s)
- Lia Moshkanbaryans
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia.
| | - Ling-Shan Chan
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia.
| | - Mark E Graham
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2145, Australia.
| |
Collapse
|
47
|
Önel SF, Rust MB, Jacob R, Renkawitz-Pohl R. Tethering membrane fusion: common and different players in myoblasts and at the synapse. J Neurogenet 2014; 28:302-15. [PMID: 24957080 PMCID: PMC4245166 DOI: 10.3109/01677063.2014.936014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Drosophila Membrane fusion is essential for the communication of membrane-defined compartments, development of multicellular organisms and tissue homeostasis. Although membrane fusion has been studied extensively, still little is known about the molecular mechanisms. Especially the intercellular fusion of cells during development and tissue homeostasis is poorly understood. Somatic muscle formation in Drosophila depends on the intercellular fusion of myoblasts. In this process, myoblasts recognize each other and adhere, thereby triggering a protein machinery that leads to electron-dense plaques, vesicles and F-actin formation at apposing membranes. Two models of how local membrane stress is achieved to induce the merging of the myoblast membranes have been proposed: the electron-dense vesicles transport and release a fusogen and F-actin bends the plasma membrane. In this review, we highlight cell-adhesion molecules and intracellular proteins known to be involved in myoblast fusion. The cell-adhesion proteins also mediate the recognition and adhesion of other cell types, such as neurons that communicate with each other via special intercellular junctions, termed chemical synapses. At these synapses, neurotransmitters are released through the intracellular fusion of synaptic vesicles with the plasma membrane. As the targeting of electron-dense vesicles in myoblasts shares some similarities with the targeting of synaptic vesicle fusion, we compare molecules required for synaptic vesicle fusion to recently identified molecules involved in myoblast fusion.
Collapse
Affiliation(s)
- Susanne Filiz Önel
- Developmental Biology, Philipps University of Marburg , 35043 Marburg , Germany
| | | | | | | |
Collapse
|
48
|
Wu Y, O'Toole ET, Girard M, Ritter B, Messa M, Liu X, McPherson PS, Ferguson SM, De Camilli P. A dynamin 1-, dynamin 3- and clathrin-independent pathway of synaptic vesicle recycling mediated by bulk endocytosis. eLife 2014; 3:e01621. [PMID: 24963135 PMCID: PMC4107917 DOI: 10.7554/elife.01621] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The exocytosis of synaptic vesicles (SVs) elicited by potent stimulation is rapidly compensated by bulk endocytosis of SV membranes leading to large endocytic vacuoles (‘bulk’ endosomes). Subsequently, these vacuoles disappear in parallel with the reappearance of new SVs. We have used synapses of dynamin 1 and 3 double knock-out neurons, where clathrin-mediated endocytosis (CME) is dramatically impaired, to gain insight into the poorly understood mechanisms underlying this process. Massive formation of bulk endosomes was not defective, but rather enhanced, in the absence of dynamin 1 and 3. The subsequent conversion of bulk endosomes into SVs was not accompanied by the accumulation of clathrin coated buds on their surface and this process proceeded even after further clathrin knock-down, suggesting its independence of clathrin. These findings support the existence of a pathway for SV reformation that bypasses the requirement for clathrin and dynamin 1/3 and that operates during intense synaptic activity. DOI:http://dx.doi.org/10.7554/eLife.01621.001 Neurons propagate electrical signals from one cell to the next using small molecules called neurotransmitters. These molecules are held inside small compartments called synaptic vesicles. Once a neuron receives an electrical stimulus, the membranes that enclose the synaptic vesicles fuse with the plasma membrane that encloses the neuron. This releases the neurotransmitters, which then trigger an electrical signal in the neighboring cell. Once the neurotransmitters are released, the vesicle membrane is rapidly reinternalized from the plasma membrane in a process called endocytosis and then recycled, ready for the next round of signal transmission. The process of synaptic vesicle membrane endocytosis and recycling has been studied extensively, and several different mechanisms by which it occurs have been identified. The best understood relies on a protein called clathrin, and is thought to be essential for nervous system function. Recently, however, a mechanism of vesicle membrane endocytosis that does not involve clathrin was identified. This mechanism, called bulk endocytosis, involves reinternalizing large regions of the cell plasma membrane to generate large compartments called vacuoles, from which new synaptic vesicles eventually form. This mechanism has been observed when neurons fire at high frequency. The cellular processes underlying bulk endocytosis are not well understood, although several studies suggest proteins called dynamins are important. Wu et al. simulated the conditions a cell experiences during high levels of activity in neurons that lacked the two major dynamins present at the synapses between neurons—dynamin 1 and dynamin 3. In these neurons, robust bulk endocytosis occurred, suggesting that these two major neuronal dynamins do not play a role in this process. Furthermore, formation of vesicles from the vacuoles generated by bulk endocytosis appeared to be clathrin-independent. These findings point to the occurrence of a pathway of synaptic vesicle recycling that bypasses the need for dynamin 1 and 3 as well as for clathrin. To reconcile these results with previously published work, Wu et al. propose that dynamins may only be required for processes that also require clathrin. But how are vesicles recycled during bulk endocytosis if dynamins are not involved? There are currently few leads to base alternative mechanisms on. Further work is required to unravel this mystery, and to provide insights into how clathrin-dependent and independent recycling processes are linked during high neuronal activity. DOI:http://dx.doi.org/10.7554/eLife.01621.002
Collapse
Affiliation(s)
- Yumei Wu
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, United States Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Eileen T O'Toole
- Department of MCD Biology, University of Colorado, Boulder, United States
| | - Martine Girard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Brigitte Ritter
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Mirko Messa
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, United States Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Shawn M Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, United States
| | - Pietro De Camilli
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, United States Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
49
|
Kirchhausen T, Owen D, Harrison SC. Molecular structure, function, and dynamics of clathrin-mediated membrane traffic. Cold Spring Harb Perspect Biol 2014; 6:a016725. [PMID: 24789820 DOI: 10.1101/cshperspect.a016725] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clathrin is a molecular scaffold for vesicular uptake of cargo at the plasma membrane, where its assembly into cage-like lattices underlies the clathrin-coated pits of classical endocytosis. This review describes the structures of clathrin, major cargo adaptors, and other proteins that participate in forming a clathrin-coated pit, loading its contents, pinching off the membrane as a lattice-enclosed vesicle, and recycling the components. It integrates as much of the structural information as possible at the time of writing into a sketch of the principal steps in coated-pit and coated-vesicle formation.
Collapse
Affiliation(s)
- Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School/PCMM, Boston, Massachusetts 02115
| | | | | |
Collapse
|
50
|
Kasprowicz J, Kuenen S, Swerts J, Miskiewicz K, Verstreken P. Dynamin photoinactivation blocks Clathrin and α-adaptin recruitment and induces bulk membrane retrieval. ACTA ACUST UNITED AC 2014; 204:1141-56. [PMID: 24662566 PMCID: PMC3971740 DOI: 10.1083/jcb.201310090] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Drosophila Dynamin prevents bulk membrane endocytosis through effects on AP2- and Clathrin-mediated stabilization of endocytic pits. Dynamin is a well-known regulator of synaptic endocytosis. Temperature-sensitive dynamin (shits1) mutations in Drosophila melanogaster or deletion of some of the mammalian Dynamins causes the accumulation of invaginated endocytic pits at synapses, sometimes also on bulk endosomes, indicating impaired membrane scission. However, complete loss of dynamin function has not been studied in neurons in vivo, and whether Dynamin acts in different aspects of synaptic vesicle formation remains enigmatic. We used acute photoinactivation and found that loss of Dynamin function blocked membrane recycling and caused the buildup of huge membrane-connected cisternae, in contrast to the invaginated pits that accumulate in shits1 mutants. Moreover, photoinactivation of Dynamin in shits1 animals converted these pits into bulk cisternae. Bulk membrane retrieval has also been seen upon Clathrin photoinactivation, and superresolution imaging indicated that acute Dynamin photoinactivation blocked Clathrin and α-adaptin relocalization to synaptic membranes upon nerve stimulation. Hence, our data indicate that Dynamin is critically involved in the stabilization of Clathrin- and AP2-dependent endocytic pits.
Collapse
Affiliation(s)
- Jaroslaw Kasprowicz
- VIB Center for the Biology of Disease, 2 Laboratory of Neuronal Communication, Department for Human Genetics, and 3 Leuven Institute for Neurodegenerative Diseases, KU Leuven, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|