1
|
Preobraschenski J, Kreutzberger AJB, Ganzella M, Münster-Wandowski A, Kreutzberger MAB, Oolsthorn LHM, Seibert S, Kiessling V, Riedel D, Witkowska A, Ahnert-Hilger G, Tamm LK, Jahn R. Synaptophysin accelerates synaptic vesicle fusion by expanding the membrane upon neurotransmitter loading. SCIENCE ADVANCES 2025; 11:eads4661. [PMID: 40267188 PMCID: PMC12017324 DOI: 10.1126/sciadv.ads4661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
Synaptic transmission is mediated by the exocytotic release of neurotransmitters stored in synaptic vesicles (SVs). SVs filled with neurotransmitters preferentially undergo exocytosis, but it is unclear how this is achieved. Here, we show that during transmitter loading, SVs substantially increase in size, which is reversible and requires synaptophysin, an abundant membrane protein with an unclear function. SVs are larger when synaptophysin is knocked out, and conversely, liposomes are smaller when reconstituted with synaptophysin. Moreover, transmitter loading of SVs accelerates fusion in vitro, which is abolished when synaptophysin is lacking despite near normal transmitter uptake. We conclude that synaptophysin functions as a curvature-promoting entity in the SV membrane, allowing for major lateral expansion of the SV membrane during neurotransmitter filling, thus increasing their propensity for exocytosis.
Collapse
Affiliation(s)
- Julia Preobraschenski
- Laboratory of Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Institute for Auditory Neuroscience, University Medical Center, Göttingen 37075, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Goettingen, Göttingen 37075, Germany
| | - Alex J. B. Kreutzberger
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville 22903, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville 22903, VA, USA
| | - Marcelo Ganzella
- Laboratory of Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | | | - Mark A. B. Kreutzberger
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville 22903, VA 22903, USA
| | - Linda H. M. Oolsthorn
- Laboratory of Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Sascha Seibert
- Institute für Integrative Neuroanatomy, Charité – Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Volker Kiessling
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville 22903, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville 22903, VA, USA
| | - Dietmar Riedel
- Facility for Transmission Electron Microscopy, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Agata Witkowska
- Laboratory of Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Gudrun Ahnert-Hilger
- Laboratory of Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Institute für Integrative Neuroanatomy, Charité – Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Lukas K. Tamm
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville 22903, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville 22903, VA, USA
| | - Reinhard Jahn
- Laboratory of Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| |
Collapse
|
2
|
Lützkendorf J, Matkovic-Rachid T, Liu S, Götz T, Gao L, Turrel O, Maglione M, Grieger M, Putignano S, Ramesh N, Ghelani T, Neumann A, Gimber N, Schmoranzer J, Stawrakakis A, Brence B, Baum D, Ludwig K, Heine M, Mielke T, Liu F, Walter AM, Wahl MC, Sigrist SJ. Blobby is a synaptic active zone assembly protein required for memory in Drosophila. Nat Commun 2025; 16:271. [PMID: 39747038 PMCID: PMC11696761 DOI: 10.1038/s41467-024-55382-9] [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: 03/27/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
At presynaptic active zones (AZs), scaffold proteins are critical for coordinating synaptic vesicle release and forming essential nanoarchitectures. However, regulatory principles steering AZ scaffold assembly, function, and plasticity remain insufficiently understood. We here identify an additional Drosophila AZ protein, "Blobby", essential for proper AZ nano-organization. Blobby biochemically associates with the ELKS family AZ scaffold protein Bruchpilot (BRP) and integrates into newly forming AZs. Loss of Blobby results in fewer AZs forming, ectopic AZ scaffold protein accumulations ("blobs") and disrupts nanoscale architecture of the BRP-AZ scaffold. Functionally, blobby mutants show diminished evoked synaptic currents due to reduced synaptic vesicle release probability and fewer functional release sites. Blobby is also present in adult brain synapses, and post-developmental knockdown of Blobby in the mushroom body impairs olfactory aversive memory consolidation. Thus, our analysis identifies an additional layer of AZ regulation critical for developmental AZ assembly but also for AZ-mediated plasticity controlling behavior.
Collapse
Affiliation(s)
- J Lützkendorf
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Matkovic-Rachid
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - T Götz
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - L Gao
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - O Turrel
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - M Maglione
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, SupraFAB, Berlin, Germany
| | - M Grieger
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Putignano
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Ramesh
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Ghelani
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A Neumann
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Gimber
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - J Schmoranzer
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - A Stawrakakis
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - B Brence
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - D Baum
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - Kai Ludwig
- Freie Universität Berlin, Institut für Chemie and Biochemie, Forschungszentrum für Elektronenmikroskopie, Berlin, Germany
| | - M Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Service Group, Berlin, Germany
| | - F Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
- University of Copenhagen, Department of Neuroscience, Copenhagen, Denmark
| | - M C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - S J Sigrist
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany.
- Charité Universitätsmedizin, NeuroCure Cluster of Excellence, Charitéplatz, Berlin, Germany.
| |
Collapse
|
3
|
Park D, Fujise K, Wu Y, Luján R, Del Olmo-Cabrera S, Wesseling JF, De Camilli P. Overlapping role of synaptophysin and synaptogyrin family proteins in determining the small size of synaptic vesicles. Proc Natl Acad Sci U S A 2024; 121:e2409605121. [PMID: 38985768 PMCID: PMC11260120 DOI: 10.1073/pnas.2409605121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
Members of the synaptophysin and synaptogyrin family are vesicle proteins with four transmembrane domains. In spite of their abundance in synaptic vesicle (SV) membranes, their role remains elusive and only mild defects at the cellular and organismal level are observed in mice lacking one or more family members. Here, we show that coexpression with synapsin in fibroblasts of each of the four brain-enriched members of this family-synaptophysin, synaptoporin, synaptogyrin 1, and synaptogyrin 3-is sufficient to generate clusters of small vesicles in the same size range of SVs. Moreover, mice lacking all these four proteins have larger SVs. We conclude that synaptophysin and synaptogyrin family proteins play an overlapping function in the biogenesis of SVs and in determining their small size.
Collapse
Affiliation(s)
- Daehun Park
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon14662, South Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon14662, South Korea
| | - Kenshiro Fujise
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
| | - Yumei Wu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
| | - Rafael Luján
- Synaptic Structure Laboratory, Departamento de Ciencias Médicas, Instituto de Biomedicina de la Universidad de Castilla-La Mancha, Facultad de Medicina, University of Castilla-La Mancha, Albacete02006, Spain
| | - Sergio Del Olmo-Cabrera
- Institute for Neurosciences Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante03550, Spain
| | - John F. Wesseling
- Institute for Neurosciences Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante03550, Spain
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
| |
Collapse
|
4
|
Yu T, Flores-Solis D, Eastep GN, Becker S, Zweckstetter M. Phosphatidylserine-dependent structure of synaptogyrin remodels the synaptic vesicle membrane. Nat Struct Mol Biol 2023:10.1038/s41594-023-01004-9. [PMID: 37217654 DOI: 10.1038/s41594-023-01004-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 04/21/2023] [Indexed: 05/24/2023]
Abstract
Synaptic vesicles are small membrane-enclosed organelles that store neurotransmitters at presynaptic terminals. The uniform morphology of synaptic vesicles is important for brain function, because it enables the storage of well-defined amounts of neurotransmitters and thus reliable synaptic transmission. Here, we show that the synaptic vesicle membrane protein synaptogyrin cooperates with the lipid phosphatidylserine to remodel the synaptic vesicle membrane. Using NMR spectroscopy, we determine the high-resolution structure of synaptogyrin and identify specific binding sites for phosphatidylserine. We further show that phosphatidylserine binding changes the transmembrane structure of synaptogyrin and is critical for membrane bending and the formation of small vesicles. Cooperative binding of phosphatidylserine to both a cytoplasmic and intravesicular lysine-arginine cluster in synaptogyrin is required for the formation of small vesicles. Together with other synaptic vesicle proteins, synaptogyrin thus can sculpt the membrane of synaptic vesicles.
Collapse
Affiliation(s)
- Taekyung Yu
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | | | - Gunnar N Eastep
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Stefan Becker
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
5
|
Sansevrino R, Hoffmann C, Milovanovic D. Condensate biology of synaptic vesicle clusters. Trends Neurosci 2023; 46:293-306. [PMID: 36725404 DOI: 10.1016/j.tins.2023.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Neuronal communication crucially relies on exocytosis of neurotransmitters from synaptic vesicles (SVs) which are clustered at synapses. To ensure reliable neurotransmitter release, synapses need to maintain an adequate pool of SVs at all times. Decades of research have established that SVs are clustered by synapsin 1, an abundant SV-associated phosphoprotein. The classical view postulates that SVs are crosslinked in a scaffold of protein-protein interactions between synapsins and their binding partners. Recent studies have shown that synapsins cluster SVs via liquid-liquid phase separation (LLPS), thus providing a new framework for the organization of the synapse. We discuss the evidence for phase separation of SVs, emphasizing emerging questions related to its regulation, specificity, and reversibility.
Collapse
Affiliation(s)
- Roberto Sansevrino
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany.
| |
Collapse
|
6
|
Praschberger R, Kuenen S, Schoovaerts N, Kaempf N, Singh J, Janssens J, Swerts J, Nachman E, Calatayud C, Aerts S, Poovathingal S, Verstreken P. Neuronal identity defines α-synuclein and tau toxicity. Neuron 2023; 111:1577-1590.e11. [PMID: 36948206 DOI: 10.1016/j.neuron.2023.02.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/22/2022] [Accepted: 02/23/2023] [Indexed: 03/24/2023]
Abstract
Pathogenic α-synuclein and tau are critical drivers of neurodegeneration, and their mutations cause neuronal loss in patients. Whether the underlying preferential neuronal vulnerability is a cell-type-intrinsic property or a consequence of increased expression levels remains elusive. Here, we explore cell-type-specific α-synuclein and tau expression in human brain datasets and use deep phenotyping as well as brain-wide single-cell RNA sequencing of >200 live neuron types in fruit flies to determine which cellular environments react most to α-synuclein or tau toxicity. We detect phenotypic and transcriptomic evidence of differential neuronal vulnerability independent of α-synuclein or tau expression levels. Comparing vulnerable with resilient neurons in Drosophila enabled us to predict numerous human neuron subtypes with increased intrinsic susceptibility to pathogenic α-synuclein or tau. By uncovering synapse- and Ca2+ homeostasis-related genes as tau toxicity modifiers, our work paves the way to leverage neuronal identity to uncover modifiers of neurodegeneration-associated toxic proteins.
Collapse
Affiliation(s)
- Roman Praschberger
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| | - Sabine Kuenen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Nils Schoovaerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Natalie Kaempf
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Jeevanjot Singh
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Jasper Janssens
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Human Genetics, 3000 Leuven, Belgium
| | - Jef Swerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Eliana Nachman
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Carles Calatayud
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Stein Aerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Human Genetics, 3000 Leuven, Belgium
| | | | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
7
|
Leahy SN, Song C, Vita DJ, Broadie K. FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 2023; 21:e3001969. [PMID: 36701299 PMCID: PMC9879533 DOI: 10.1371/journal.pbio.3001969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Noonan syndrome (NS) and NS with multiple lentigines (NSML) cognitive dysfunction are linked to SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) gain-of-function (GoF) and loss-of-function (LoF), respectively. In Drosophila disease models, we find both SHP2 mutations from human patients and corkscrew (csw) homolog LoF/GoF elevate glutamatergic transmission. Cell-targeted RNAi and neurotransmitter release analyses reveal a presynaptic requirement. Consistently, all mutants exhibit reduced synaptic depression during high-frequency stimulation. Both LoF and GoF mutants also show impaired synaptic plasticity, including reduced facilitation, augmentation, and post-tetanic potentiation. NS/NSML diseases are characterized by elevated MAPK/ERK signaling, and drugs suppressing this signaling restore normal neurotransmission in mutants. Fragile X syndrome (FXS) is likewise characterized by elevated MAPK/ERK signaling. Fragile X Mental Retardation Protein (FMRP) binds csw mRNA and neuronal Csw protein is elevated in Drosophila fragile X mental retardation 1 (dfmr1) nulls. Moreover, phosphorylated ERK (pERK) is increased in dfmr1 and csw null presynaptic boutons. We find presynaptic pERK activation in response to stimulation is reduced in dfmr1 and csw nulls. Trans-heterozygous csw/+; dfmr1/+ recapitulate elevated presynaptic pERK activation and function, showing FMRP and Csw/SHP2 act within the same signaling pathway. Thus, a FMRP and SHP2 MAPK/ERK regulative mechanism controls basal and activity-dependent neurotransmission strength.
Collapse
Affiliation(s)
- Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Dominic J. Vita
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
8
|
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: 38] [Impact Index Per Article: 9.5] [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
|
9
|
Bhimreddy M, Rushton E, Kopke DL, Broadie K. Secreted C-type lectin regulation of neuromuscular junction synaptic vesicle dynamics modulates coordinated movement. J Cell Sci 2021; 134:261954. [PMID: 33973638 DOI: 10.1242/jcs.257592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/03/2021] [Indexed: 11/20/2022] Open
Abstract
The synaptic cleft manifests enriched glycosylation, with structured glycans coordinating signaling between presynaptic and postsynaptic cells. Glycosylated signaling ligands orchestrating communication are tightly regulated by secreted glycan-binding lectins. Using the Drosophila neuromuscular junction (NMJ) as a model glutamatergic synapse, we identify a new Ca2+-binding (C-type) lectin, Lectin-galC1 (LGC1), which modulates presynaptic function and neurotransmission strength. We find that LGC1 is enriched in motoneuron presynaptic boutons and secreted into the NMJ extracellular synaptomatrix. We show that LGC1 limits locomotor peristalsis and coordinated movement speed, with a specific requirement for synaptic function, but not NMJ architecture. LGC1 controls neurotransmission strength by limiting presynaptic active zone (AZ) and postsynaptic glutamate receptor (GluR) aligned synapse number, reducing both spontaneous and stimulation-evoked synaptic vesicle (SV) release, and capping SV cycling rate. During high-frequency stimulation (HFS), mutants have faster synaptic depression and impaired recovery while replenishing depleted SV pools. Although LGC1 removal increases the number of glutamatergic synapses, we find that LGC1-null mutants exhibit decreased SV density within presynaptic boutons, particularly SV pools at presynaptic active zones. Thus, LGC1 regulates NMJ neurotransmission to modulate coordinated movement.
Collapse
Affiliation(s)
- Meghana Bhimreddy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Emma Rushton
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Danielle L Kopke
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA.,Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA.,Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| |
Collapse
|
10
|
Hintze A, Gültas M, Semmelhack EA, Wichmann C. Ultrastructural maturation of the endbulb of Held active zones comparing wild-type and otoferlin-deficient mice. iScience 2021; 24:102282. [PMID: 33851098 PMCID: PMC8022229 DOI: 10.1016/j.isci.2021.102282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/18/2021] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Endbulbs of Held are located in the anteroventral cochlear nucleus and present the first central synapses of the auditory pathway. During development, endbulbs mature functionally to enable rapid and powerful synaptic transmission with high temporal precision. This process is accompanied by morphological changes of endbulb terminals. Loss of the hair cell-specific protein otoferlin (Otof) abolishes neurotransmission in the cochlea and results in the smaller endbulb of Held terminals. Thus, peripheral hearing impairment likely also leads to alterations in the morphological synaptic vesicle (SV) pool size at individual endbulb of Held active zones (AZs). Here, we investigated endbulb AZs in pre-hearing, young, and adult wild-type and Otof−/− mice. During maturation, SV numbers at endbulb AZs increased in wild-type mice but were found to be reduced in Otof−/− mice. The SV population at a distance of 0–15 nm was most strongly affected. Finally, overall SV diameters decreased in Otof−/− animals during maturation. Maturation of wt endbulb of Held active zones leads to more synaptic vesicles At endbulbs of otoferlin knockout mice, synaptic vesicles decline with age Mainly two distinct synaptic vesicle populations are affected Synaptic vesicles sizes are reduced in six-month-old otoferlin knockout animals
Collapse
Affiliation(s)
- Anika Hintze
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.,Collaborative Research Center 1286, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Sciences, Georg-August-University Göttingen, Göttingen, Germany
| | - Esther A Semmelhack
- Developmental, Neural, and Behavioral Biology MSc/PhD Program, 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, 37075 Göttingen, Germany.,Collaborative Research Center 1286, University of Göttingen, Göttingen, Germany
| |
Collapse
|
11
|
Hartwig C, Méndez GM, Bhattacharjee S, Vrailas-Mortimer AD, Zlatic SA, Freeman AAH, Gokhale A, Concilli M, Werner E, Sapp Savas C, Rudin-Rush S, Palmer L, Shearing N, Margewich L, McArthy J, Taylor S, Roberts B, Lupashin V, Polishchuk RS, Cox DN, Jorquera RA, Faundez V. Golgi-Dependent Copper Homeostasis Sustains Synaptic Development and Mitochondrial Content. J Neurosci 2021; 41:215-233. [PMID: 33208468 PMCID: PMC7810662 DOI: 10.1523/jneurosci.1284-20.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/02/2020] [Accepted: 11/09/2020] [Indexed: 01/05/2023] Open
Abstract
Rare genetic diseases preponderantly affect the nervous system causing neurodegeneration to neurodevelopmental disorders. This is the case for both Menkes and Wilson disease, arising from mutations in ATP7A and ATP7B, respectively. The ATP7A and ATP7B proteins localize to the Golgi and regulate copper homeostasis. We demonstrate genetic and biochemical interactions between ATP7 paralogs with the conserved oligomeric Golgi (COG) complex, a Golgi apparatus vesicular tether. Disruption of Drosophila copper homeostasis by ATP7 tissue-specific transgenic expression caused alterations in epidermis, aminergic, sensory, and motor neurons. Prominent among neuronal phenotypes was a decreased mitochondrial content at synapses, a phenotype that paralleled with alterations of synaptic morphology, transmission, and plasticity. These neuronal and synaptic phenotypes caused by transgenic expression of ATP7 were rescued by downregulation of COG complex subunits. We conclude that the integrity of Golgi-dependent copper homeostasis mechanisms, requiring ATP7 and COG, are necessary to maintain mitochondria functional integrity and localization to synapses.SIGNIFICANCE STATEMENT Menkes and Wilson disease affect copper homeostasis and characteristically afflict the nervous system. However, their molecular neuropathology mechanisms remain mostly unexplored. We demonstrate that copper homeostasis in neurons is maintained by two factors that localize to the Golgi apparatus, ATP7 and the conserved oligomeric Golgi (COG) complex. Disruption of these mechanisms affect mitochondrial function and localization to synapses as well as neurotransmission and synaptic plasticity. These findings suggest communication between the Golgi apparatus and mitochondria through homeostatically controlled cellular copper levels and copper-dependent enzymatic activities in both organelles.
Collapse
Affiliation(s)
- Cortnie Hartwig
- Departments of Cell Biology, Emory University, Atlanta, Georgia 30322
| | | | - Shatabdi Bhattacharjee
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | | | | | - Amanda A H Freeman
- The Center for the Study of Human Health, Emory University, Atlanta, Georgia 30322
| | - Avanti Gokhale
- Departments of Cell Biology, Emory University, Atlanta, Georgia 30322
| | - Mafalda Concilli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Erica Werner
- Departments of Cell Biology, Emory University, Atlanta, Georgia 30322
| | | | | | - Laura Palmer
- Departments of Cell Biology, Emory University, Atlanta, Georgia 30322
| | - Nicole Shearing
- Departments of Cell Biology, Emory University, Atlanta, Georgia 30322
| | - Lindsey Margewich
- School of Biological Sciences, IL State University, Normal, Illinois 617901
| | - Jacob McArthy
- School of Biological Sciences, IL State University, Normal, Illinois 617901
| | - Savanah Taylor
- School of Biological Sciences, IL State University, Normal, Illinois 617901
| | - Blaine Roberts
- Departments of Biochemistry, Emory University, Atlanta, Georgia 30322
| | - Vladimir Lupashin
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Roman S Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy
| | - Daniel N Cox
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Ramon A Jorquera
- Neuroscience Department, Universidad Central del Caribe, Bayamon, Puerto Rico 00956
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370186, Chile
| | - Victor Faundez
- Departments of Cell Biology, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
12
|
Li TN, Chen YJ, Lu TY, Wang YT, Lin HC, Yao CK. A positive feedback loop between Flower and PI(4,5)P 2 at periactive zones controls bulk endocytosis in Drosophila. eLife 2020; 9:60125. [PMID: 33300871 PMCID: PMC7748424 DOI: 10.7554/elife.60125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/09/2020] [Indexed: 01/01/2023] Open
Abstract
Synaptic vesicle (SV) endocytosis is coupled to exocytosis to maintain SV pool size and thus neurotransmitter release. Intense stimulation induces activity-dependent bulk endocytosis (ADBE) to recapture large quantities of SV constituents in large endosomes from which SVs reform. How these consecutive processes are spatiotemporally coordinated remains unknown. Here, we show that Flower Ca2+ channel-dependent phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) compartmentalization governs control of these processes in Drosophila. Strong stimuli trigger PI(4,5)P2 microdomain formation at periactive zones. Upon exocytosis, Flower translocates from SVs to periactive zones, where it increases PI(4,5)P2 levels via Ca2+ influxes. Remarkably, PI(4,5)P2 directly enhances Flower channel activity, thereby establishing a positive feedback loop for PI(4,5)P2 microdomain compartmentalization. PI(4,5)P2 microdomains drive ADBE and SV reformation from bulk endosomes. PI(4,5)P2 further retrieves Flower to bulk endosomes, terminating endocytosis. We propose that the interplay between Flower and PI(4,5)P2 is the crucial spatiotemporal cue that couples exocytosis to ADBE and subsequent SV reformation.
Collapse
Affiliation(s)
- Tsai-Ning Li
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yu-Jung Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ting-Yi Lu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - You-Tung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hsin-Chieh Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chi-Kuang Yao
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
13
|
Ariotti N, Wu Y, Okano S, Gambin Y, Follett J, Rae J, Ferguson C, Teasdale RD, Alexandrov K, Meunier FA, Hill MM, Parton RG. An inverted CAV1 (caveolin 1) topology defines novel autophagy-dependent exosome secretion from prostate cancer cells. Autophagy 2020; 17:2200-2216. [DOI: 10.1080/15548627.2020.1820787] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Nicholas Ariotti
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Mark Wainwright Analytical Centre, Electron Microscope Unit, The University of New South Wales, Sydney, Australia
| | - Yeping Wu
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Satomi Okano
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Yann Gambin
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jordan Follett
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - James Rae
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Charles Ferguson
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Rohan D. Teasdale
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Kirill Alexandrov
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Frederic A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Michelle M. Hill
- UQ Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Robert G. Parton
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- The Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Australia
| |
Collapse
|
14
|
Rodriguez-López J, Arrojo M, Paz E, Páramo M, Costas J. Identification of relevant hub genes for early intervention at gene coexpression modules with altered predicted expression in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2020; 98:109815. [PMID: 31715283 DOI: 10.1016/j.pnpbp.2019.109815] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/25/2019] [Accepted: 11/08/2019] [Indexed: 01/20/2023]
Abstract
Genetic risk for schizophrenia is due to the joint effect of multiple genes acting mainly at two different processes, prenatal/perinatal neurodevelopment and adolescence/early adulthood synapse maturation. Identification of important genes at the second process is of relevance for early intervention. The aim of this work was to identify gene co-expression modules with altered expression in schizophrenia during adolescence/early adulthood. To this goal, we predicted frontal cortex gene expression in one discovery sample, the largest GWAS of schizophrenia from the Psychiatric Genomics Consortium, using S-prediXcan, and in one target sample, consisting of 625 schizophrenic patients and 819 controls from Spain, using prediXcan. Prediction models were trained on GTEx frontal cortex expression dataset. In parallel, we identified brain co-expression modules from BrainSpan using WGCNA. Then, we estimated polygenic risk scores based on predicted expression (PE-PRS) for each co-expression module in the target sample, based on PE-PRS model from the discovery sample. This analysis led to the identification of a module with mainly adolescence/adulthood expression whose PE-PRS was significantly associated with schizophrenia. The module was significantly enriched in synaptic processes. Several hub genes at this module are drugabble, according to the drug-gene interaction database, and/or involved in synaptic transmission, such as the voltage-gated ion channels SCN2B and KCNAB2, the calcium calmodulin kinases CAMK2A and CAMK1G, or genes involved in synaptic vesicle cycle, such as DNM1, or SYNGR1. Therefore, identification of this module may be the first step in patient stratification based on biology, as well as in drug design and drug repurposing efforts.
Collapse
Affiliation(s)
- Julio Rodriguez-López
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Arrojo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Eduardo Paz
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Mario Páramo
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain; Servizo de Psiquiatría, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain
| | - Javier Costas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, Spain.
| |
Collapse
|
15
|
Haytural H, Mermelekas G, Emre C, Nigam SM, Carroll SL, Winblad B, Bogdanovic N, Barthet G, Granholm AC, Orre LM, Tjernberg LO, Frykman S. The Proteome of the Dentate Terminal Zone of the Perforant Path Indicates Presynaptic Impairment in Alzheimer Disease. Mol Cell Proteomics 2020; 19:128-141. [PMID: 31699905 PMCID: PMC6944231 DOI: 10.1074/mcp.ra119.001737] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/05/2019] [Indexed: 01/13/2023] Open
Abstract
Synaptic dysfunction is an early pathogenic event in Alzheimer disease (AD) that contributes to network disturbances and cognitive decline. Some synapses are more vulnerable than others, including the synapses of the perforant path, which provides the main excitatory input to the hippocampus. To elucidate the molecular mechanisms underlying the dysfunction of these synapses, we performed an explorative proteomic study of the dentate terminal zone of the perforant path. The outer two-thirds of the molecular layer of the dentate gyrus, where the perforant path synapses are located, was microdissected from five subjects with AD and five controls. The microdissected tissues were dissolved and digested by trypsin. Peptides from each sample were labeled with different isobaric tags, pooled together and pre-fractionated into 72 fractions by high-resolution isoelectric focusing. Each fraction was then analyzed by liquid chromatography-mass spectrometry. We quantified the relative expression levels of 7322 proteins, whereof 724 showed significantly altered levels in AD. Our comprehensive data analysis using enrichment and pathway analyses strongly indicated that presynaptic signaling, such as exocytosis and synaptic vesicle cycle processes, is severely disturbed in this area in AD, whereas postsynaptic proteins remained unchanged. Among the significantly altered proteins, we selected three of the most downregulated synaptic proteins; complexin-1, complexin-2 and synaptogyrin-1, for further validation, using a new cohort consisting of six AD and eight control cases. Semi-quantitative analysis of immunohistochemical staining confirmed decreased levels of complexin-1, complexin-2 and synaptogyrin-1 in the outer two-thirds of the molecular layer of the dentate gyrus in AD. Our in-depth proteomic analysis provides extensive knowledge on the potential molecular mechanism underlying synaptic dysfunction related to AD and supports that presynaptic alterations are more important than postsynaptic changes in early stages of the disease. The specific synaptic proteins identified could potentially be targeted to halt synaptic dysfunction in AD.
Collapse
Affiliation(s)
- Hazal Haytural
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.
| | - Georgios Mermelekas
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ceren Emre
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | | | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Bengt Winblad
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden; Karolinska University Hospital, Theme Aging, Stockholm, Sweden
| | - Nenad Bogdanovic
- Karolinska University Hospital, Theme Aging, Stockholm, Sweden; Division of Clinical geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Gaël Barthet
- Interdisciplinary Institute for Neuroscience, CNRS UMR, Bordeaux, France; University of Bordeaux, Bordeaux, France
| | - Ann-Charlotte Granholm
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden; Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado
| | - Lukas M Orre
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Susanne Frykman
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
16
|
Two Pathways for the Activity-Dependent Growth and Differentiation of Synaptic Boutons in Drosophila. eNeuro 2019; 6:ENEURO.0060-19.2019. [PMID: 31387877 PMCID: PMC6709223 DOI: 10.1523/eneuro.0060-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/15/2019] [Accepted: 07/26/2019] [Indexed: 11/21/2022] Open
Abstract
Synapse formation can be promoted by intense activity. At the Drosophila larval neuromuscular junction (NMJ), new synaptic boutons can grow acutely in response to patterned stimulation. We combined confocal imaging with electron microscopy and tomography to investigate the initial stages of growth and differentiation of new presynaptic boutons at the Drosophila NMJ. We found that the new boutons can form rapidly in intact larva in response to intense crawling activity, and we observed two different patterns of bouton formation and maturation. The first pathway involves the growth of filopodia followed by a formation of boutons that are initially devoid of synaptic vesicles (SVs) but filled with filamentous matrix. The second pathway involves rapid budding of synaptic boutons packed with SVs, and these more mature boutons are sometimes capable of exocytosis/endocytosis. We demonstrated that intense activity predominantly promotes the second pathway, i.e., budding of more mature boutons filled with SVs. We also showed that this pathway depends on synapsin (Syn), a neuronal protein which reversibly associates with SVs and mediates their clustering via a protein kinase A (PKA)-dependent mechanism. Finally, we took advantage of the temperature-sensitive mutant sei to demonstrate that seizure activity can promote very rapid budding of new boutons filled with SVs, and this process occurs at scale of minutes. Altogether, these results demonstrate that intense activity acutely and selectively promotes rapid budding of new relatively mature presynaptic boutons filled with SVs, and that this process is regulated via a PKA/Syn-dependent pathway.
Collapse
|
17
|
Raja MK, Preobraschenski J, Del Olmo-Cabrera S, Martinez-Turrillas R, Jahn R, Perez-Otano I, Wesseling JF. Elevated synaptic vesicle release probability in synaptophysin/gyrin family quadruple knockouts. eLife 2019; 8:40744. [PMID: 31090538 PMCID: PMC6519982 DOI: 10.7554/elife.40744] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 04/18/2019] [Indexed: 01/05/2023] Open
Abstract
Synaptophysins 1 and 2 and synaptogyrins 1 and 3 constitute a major family of synaptic vesicle membrane proteins. Unlike other widely expressed synaptic vesicle proteins such as vSNAREs and synaptotagmins, the primary function has not been resolved. Here, we report robust elevation in the probability of release of readily releasable vesicles with both high and low release probabilities at a variety of synapse types from knockout mice missing all four family members. Neither the number of readily releasable vesicles, nor the timing of recruitment to the readily releasable pool was affected. The results suggest that family members serve as negative regulators of neurotransmission, acting directly at the level of exocytosis to dampen connection strength selectively when presynaptic action potentials fire at low frequency. The widespread expression suggests that chemical synapses may play a frequency filtering role in biological computation that is more elemental than presently envisioned. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Mathan K Raja
- Department of Neuroscience, Universidad de Navarra, Pamplona, Spain
| | - Julia Preobraschenski
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | | | | | - Reinhard Jahn
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Isabel Perez-Otano
- Department of Neuroscience, Universidad de Navarra, Pamplona, Spain.,Institute for Neurosciences CSIC-UMH, San Juan de Alicante, Spain
| | - John F Wesseling
- Department of Neuroscience, Universidad de Navarra, Pamplona, Spain.,Institute for Neurosciences CSIC-UMH, San Juan de Alicante, Spain
| |
Collapse
|
18
|
The ontogeny of synaptophysin expression patterns on the GABAergic ciliary band-associated strand during larval development of the sea urchin, Hemicentrotus pulcherrimus A. Agassiz, 1864. ZOOMORPHOLOGY 2018. [DOI: 10.1007/s00435-018-0424-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
19
|
Multimodal Rehabilitation Program Promotes Motor Function Recovery of Rats After Ischemic Stroke by Upregulating Expressions of GAP-43, SYN, HSP70, and C-MYC. J Stroke Cerebrovasc Dis 2018; 27:2829-2839. [PMID: 30093210 DOI: 10.1016/j.jstrokecerebrovasdis.2018.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/30/2018] [Accepted: 06/14/2018] [Indexed: 11/21/2022] Open
|
20
|
McInnes J, Wierda K, Snellinx A, Bounti L, Wang YC, Stancu IC, Apóstolo N, Gevaert K, Dewachter I, Spires-Jones TL, De Strooper B, De Wit J, Zhou L, Verstreken P. Synaptogyrin-3 Mediates Presynaptic Dysfunction Induced by Tau. Neuron 2018; 97:823-835.e8. [DOI: 10.1016/j.neuron.2018.01.022] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/07/2017] [Accepted: 01/09/2018] [Indexed: 11/25/2022]
|
21
|
Ugur B, Bao H, Stawarski M, Duraine LR, Zuo Z, Lin YQ, Neely GG, Macleod GT, Chapman ER, Bellen HJ. The Krebs Cycle Enzyme Isocitrate Dehydrogenase 3A Couples Mitochondrial Metabolism to Synaptic Transmission. Cell Rep 2017; 21:3794-3806. [PMID: 29281828 PMCID: PMC5747319 DOI: 10.1016/j.celrep.2017.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 11/10/2017] [Accepted: 12/01/2017] [Indexed: 10/18/2022] Open
Abstract
Neurotransmission is a tightly regulated Ca2+-dependent process. Upon Ca2+ influx, Synaptotagmin1 (Syt1) promotes fusion of synaptic vesicles (SVs) with the plasma membrane. This requires regulation at multiple levels, but the role of metabolites in SV release is unclear. Here, we uncover a role for isocitrate dehydrogenase 3a (idh3a), a Krebs cycle enzyme, in neurotransmission. Loss of idh3a leads to a reduction of the metabolite, alpha-ketoglutarate (αKG), causing defects in synaptic transmission similar to the loss of syt1. Supplementing idh3a flies with αKG suppresses these defects through an ATP or neurotransmitter-independent mechanism. Indeed, αKG, but not glutamate, enhances Syt1-dependent fusion in a reconstitution assay. αKG promotes interaction between the C2-domains of Syt1 and phospholipids. The data reveal conserved metabolic regulation of synaptic transmission via αKG. Our studies provide a synaptic role for αKG, a metabolite that has been proposed as a treatment for aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Berrak Ugur
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Huan Bao
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA; Howard Hughes Medical Institute, University of Wisconsin, Madison, WI 53705, USA
| | - Michal Stawarski
- Department of Biological Sciences and Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Lita R Duraine
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Qi Lin
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - G Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Gregory T Macleod
- Department of Biological Sciences and Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA; Howard Hughes Medical Institute, University of Wisconsin, Madison, WI 53705, USA
| | - Hugo J Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
22
|
Complexin Mutants Reveal Partial Segregation between Recycling Pathways That Drive Evoked and Spontaneous Neurotransmission. J Neurosci 2017; 37:383-396. [PMID: 28077717 DOI: 10.1523/jneurosci.1854-16.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/21/2022] Open
Abstract
Synaptic vesicles fuse at morphological specializations in the presynaptic terminal termed active zones (AZs). Vesicle fusion can occur spontaneously or in response to an action potential. Following fusion, vesicles are retrieved and recycled within nerve terminals. It is still unclear whether vesicles that fuse spontaneously or following evoked release share similar recycling mechanisms. Genetic deletion of the SNARE-binding protein complexin dramatically increases spontaneous fusion, with the protein serving as the synaptic vesicle fusion clamp at Drosophila synapses. We examined synaptic vesicle recycling pathways at complexin null neuromuscular junctions, where spontaneous release is dramatically enhanced. We combined loading of the lipophilic dye FM1-43 with photoconversion, electron microscopy, and electrophysiology to monitor evoked and spontaneous recycling vesicle pools. We found that the total number of recycling vesicles was equal to those retrieved through spontaneous and evoked pools, suggesting that retrieval following fusion is partially segregated for spontaneous and evoked release. In addition, the kinetics of FM1-43 destaining and synaptic depression measured in the presence of the vesicle-refilling blocker bafilomycin indicated that spontaneous and evoked recycling pools partially intermix during the release process. Finally, FM1-43 photoconversion combined with electron microscopy analysis indicated that spontaneous recycling preferentially involves synaptic vesicles in the vicinity of AZs, whereas vesicles recycled following evoked release involve a larger intraterminal pool. Together, these results suggest that spontaneous and evoked vesicles use separable recycling pathways and then partially intermix during subsequent rounds of fusion. SIGNIFICANCE STATEMENT Neurotransmitter release involves fusion of synaptic vesicles with the plasma membrane in response to an action potential, or spontaneously in the absence of stimulation. Upon fusion, vesicles are retrieved and recycled, and it is unclear whether recycling pathways for evoked and spontaneous vesicles are segregated after fusion. We addressed this question by taking advantage of preparations lacking the synaptic protein complexin, which have elevated spontaneous release that enables reliable tracking of the spontaneous recycling pool. Our results suggest that spontaneous and evoked recycling pathways are segregated during the retrieval process but can partially intermix during stimulation.
Collapse
|
23
|
Yao CK, Liu YT, Lee IC, Wang YT, Wu PY. A Ca2+ channel differentially regulates Clathrin-mediated and activity-dependent bulk endocytosis. PLoS Biol 2017; 15:e2000931. [PMID: 28414717 PMCID: PMC5393565 DOI: 10.1371/journal.pbio.2000931] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/21/2017] [Indexed: 11/19/2022] Open
Abstract
Clathrin-mediated endocytosis (CME) and activity-dependent bulk endocytosis (ADBE) are two predominant forms of synaptic vesicle (SV) endocytosis, elicited by moderate and strong stimuli, respectively. They are tightly coupled with exocytosis for sustained neurotransmission. However, the underlying mechanisms are ill defined. We previously reported that the Flower (Fwe) Ca2+ channel present in SVs is incorporated into the periactive zone upon SV fusion, where it triggers CME, thus coupling exocytosis to CME. Here, we show that Fwe also promotes ADBE. Intriguingly, the effects of Fwe on CME and ADBE depend on the strength of the stimulus. Upon mild stimulation, Fwe controls CME independently of Ca2+ channeling. However, upon strong stimulation, Fwe triggers a Ca2+ influx that initiates ADBE. Moreover, knockout of rodent fwe in cultured rat hippocampal neurons impairs but does not completely abolish CME, similar to the loss of Drosophila fwe at the neuromuscular junction, suggesting that Fwe plays a regulatory role in regulating CME across species. In addition, the function of Fwe in ADBE is conserved at mammalian central synapses. Hence, Fwe exerts different effects in response to different stimulus strengths to control two major modes of endocytosis.
Collapse
Affiliation(s)
- Chi-Kuang Yao
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
- Neuroscience Program in Academia Sinica, Academia Sinica, Nankang, Taipei, Taiwan
- Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
- * E-mail:
| | - Yu-Tzu Liu
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - I-Chi Lee
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - You-Tung Wang
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - Ping-Yen Wu
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| |
Collapse
|
24
|
Sun Q, Qi X, Zhang Y, Wu X, Liang M, Li C, Li D, Cardona CJ, Xing Z. Synaptogyrin-2 Promotes Replication of a Novel Tick-borne Bunyavirus through Interacting with Viral Nonstructural Protein NSs. J Biol Chem 2016; 291:16138-49. [PMID: 27226560 DOI: 10.1074/jbc.m116.715599] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Indexed: 01/24/2023] Open
Abstract
Synaptogyrin-2 is a non-neuronal member of the synaptogyrin family involved in synaptic vesicle biogenesis and trafficking. Little is known about the function of synaptogyrin-2. Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease characterized by high fever, thrombocytopenia, and leukocytopenia with high mortality, caused by a novel tick-borne phlebovirus in the family Bunyaviridae. Our previous studies have shown that the viral nonstructural protein NSs forms inclusion bodies (IBs) that are involved in viral immune evasion, as well as viral RNA replication. In this study, we sought to elucidate the mechanism by which NSs formed the IBs, a lipid droplet-based structure confirmed by NSs co-localization with perilipin A and adipose differentiation-related protein (ADRP). Through a high throughput screening, we identified synaptogyrin-2 to be highly up-regulated in response to SFTS bunyavirus (SFTSV) infection and to be a promoter of viral replication. We demonstrated that synaptogyrin-2 interacted with NSs and was translocated into the IBs, which were reconstructed from lipid droplets into large structures in infection. Viral RNA replication decreased, and infectious virus titers were lowered significantly when synaptogyrin-2 was silenced in specific shRNA-expressing cells, which correlated with the reduced number of the large IBs restructured from regular lipid droplets. We hypothesize that synaptogyrin-2 is essential to promoting the formation of the IBs to become virus factories for viral RNA replication through its interaction with NSs. These findings unveil the function of synaptogyrin-2 as an enhancer in viral infection.
Collapse
Affiliation(s)
- Qiyu Sun
- From the State Key Laboratory of Pharmaceutical Biotechnology and Medical School, Nanjing University, Nanjing 210093, China
| | - Xian Qi
- the Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Yan Zhang
- From the State Key Laboratory of Pharmaceutical Biotechnology and Medical School, Nanjing University, Nanjing 210093, China
| | - Xiaodong Wu
- From the State Key Laboratory of Pharmaceutical Biotechnology and Medical School, Nanjing University, Nanjing 210093, China
| | - Mifang Liang
- the National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China, and
| | - Chuan Li
- the National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China, and
| | - Dexin Li
- the National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China, and
| | - Carol J Cardona
- the Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, St. Paul, Minnesota 55108
| | - Zheng Xing
- From the State Key Laboratory of Pharmaceutical Biotechnology and Medical School, Nanjing University, Nanjing 210093, China, the Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, St. Paul, Minnesota 55108
| |
Collapse
|
25
|
Rajappa R, Gauthier-Kemper A, Böning D, Hüve J, Klingauf J. Synaptophysin 1 Clears Synaptobrevin 2 from the Presynaptic Active Zone to Prevent Short-Term Depression. Cell Rep 2016; 14:1369-1381. [DOI: 10.1016/j.celrep.2016.01.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/23/2015] [Accepted: 01/06/2016] [Indexed: 10/22/2022] Open
|
26
|
Zhang KX, Tan L, Pellegrini M, Zipursky SL, McEwen JM. Rapid Changes in the Translatome during the Conversion of Growth Cones to Synaptic Terminals. Cell Rep 2016; 14:1258-1271. [PMID: 26832407 DOI: 10.1016/j.celrep.2015.12.102] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 12/04/2015] [Accepted: 12/21/2015] [Indexed: 12/01/2022] Open
Abstract
A common step in the formation of neural circuits is the conversion of growth cones to presynaptic terminals. Characterizing patterns of global gene expression during this process is problematic due to the cellular diversity of the brain and the complex temporal dynamics of development. Here, we take advantage of the synchronous conversion of Drosophila photoreceptor growth cones into presynaptic terminals to explore global changes in gene expression during presynaptic differentiation. Using a tandemly tagged ribosome trap (T-TRAP) and RNA sequencing (RNA-seq) at multiple developmental times, we observed dramatic changes in coding and non-coding RNAs with presynaptic differentiation. Marked changes in the mRNA encoding transmembrane and secreted proteins occurred preferentially. The 3' UTRs of transcripts encoding synaptic proteins were preferentially lengthened, and these extended UTRs were preferentially enriched for sites recognized by RNA binding proteins. These data provide a rich resource for uncovering the regulatory logic underlying presynaptic differentiation.
Collapse
Affiliation(s)
- Kelvin Xi Zhang
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA
| | - Liming Tan
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, P.O. Box 951606, Los Angeles, CA 90095, USA
| | - S Lawrence Zipursky
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA.
| | - Jason M McEwen
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA
| |
Collapse
|
27
|
Harris KP, Littleton JT. Transmission, Development, and Plasticity of Synapses. Genetics 2015; 201:345-75. [PMID: 26447126 PMCID: PMC4596655 DOI: 10.1534/genetics.115.176529] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/28/2015] [Indexed: 01/03/2023] Open
Abstract
Chemical synapses are sites of contact and information transfer between a neuron and its partner cell. Each synapse is a specialized junction, where the presynaptic cell assembles machinery for the release of neurotransmitter, and the postsynaptic cell assembles components to receive and integrate this signal. Synapses also exhibit plasticity, during which synaptic function and/or structure are modified in response to activity. With a robust panel of genetic, imaging, and electrophysiology approaches, and strong evolutionary conservation of molecular components, Drosophila has emerged as an essential model system for investigating the mechanisms underlying synaptic assembly, function, and plasticity. We will discuss techniques for studying synapses in Drosophila, with a focus on the larval neuromuscular junction (NMJ), a well-established model glutamatergic synapse. Vesicle fusion, which underlies synaptic release of neurotransmitters, has been well characterized at this synapse. In addition, studies of synaptic assembly and organization of active zones and postsynaptic densities have revealed pathways that coordinate those events across the synaptic cleft. We will also review modes of synaptic growth and plasticity at the fly NMJ, and discuss how pre- and postsynaptic cells communicate to regulate plasticity in response to activity.
Collapse
Affiliation(s)
- Kathryn P Harris
- Department of Biology and Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - J Troy Littleton
- Department of Biology and Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
28
|
Kioumourtzoglou D, Pryor PR, Gould GW, Bryant NJ. Alternative routes to the cell surface underpin insulin-regulated membrane trafficking of GLUT4. J Cell Sci 2015; 128:2423-9. [PMID: 26071524 PMCID: PMC4510850 DOI: 10.1242/jcs.166561] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 05/26/2015] [Indexed: 01/12/2023] Open
Abstract
Insulin-stimulated delivery of glucose transporters (GLUT4, also known as SLC2A4) from specialized intracellular GLUT4 storage vesicles (GSVs) to the surface of fat and muscle cells is central to whole-body glucose regulation. This translocation and subsequent internalization of GLUT4 back into intracellular stores transits through numerous small membrane-bound compartments (internal GLUT4-containing vesicles; IGVs) including GSVs, but the function of these different compartments is not clear. Cellugyrin (also known as synaptogyrin-2) and sortilin define distinct populations of IGV; sortilin-positive IGVs represent GSVs, but the function of cellugyrin-containing IGVs is unknown. Here, we demonstrate a role for cellugyrin in intracellular sequestration of GLUT4 in HeLa cells and have used a proximity ligation assay to follow changes in pairwise associations between cellugyrin, sortilin, GLUT4 and membrane trafficking machinery following insulin-stimulation of 3T3-L1 adipoctyes. Our data suggest that insulin stimulates traffic from cellugyrin-containing to sortilin-containing membranes, and that cellugyrin-containing IGVs provide an insulin-sensitive reservoir to replenish GSVs following insulin-stimulated exocytosis of GLUT4. Furthermore, our data support the existence of a pathway from cellugyrin-containing membranes to the surface of 3T3-L1 adipocytes that bypasses GSVs under basal conditions, and that insulin diverts traffic away from this into GSVs.
Collapse
Affiliation(s)
- Dimitrios Kioumourtzoglou
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK Department of Biology, University of York, York YO10 5DD, UK
| | - Paul R Pryor
- Department of Biology, University of York, York YO10 5DD, UK Centre for Immunology and Infection, Hull York Medical School, University of York, York YO10 5DD, UK
| | - Gwyn W Gould
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Nia J Bryant
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK Department of Biology, University of York, York YO10 5DD, UK
| |
Collapse
|
29
|
Burow DA, Umeh-Garcia MC, True MB, Bakhaj CD, Ardell DH, Cleary MD. Dynamic regulation of mRNA decay during neural development. Neural Dev 2015; 10:11. [PMID: 25896902 PMCID: PMC4413985 DOI: 10.1186/s13064-015-0038-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/07/2015] [Indexed: 01/06/2023] Open
Abstract
Background Gene expression patterns are determined by rates of mRNA transcription and decay. While transcription is known to regulate many developmental processes, the role of mRNA decay is less extensively defined. A critical step toward defining the role of mRNA decay in neural development is to measure genome-wide mRNA decay rates in neural tissue. Such information should reveal the degree to which mRNA decay contributes to differential gene expression and provide a foundation for identifying regulatory mechanisms that affect neural mRNA decay. Results We developed a technique that allows genome-wide mRNA decay measurements in intact Drosophila embryos, across all tissues and specifically in the nervous system. Our approach revealed neural-specific decay kinetics, including stabilization of transcripts encoding regulators of axonogenesis and destabilization of transcripts encoding ribosomal proteins and histones. We also identified correlations between mRNA stability and physiologic properties of mRNAs; mRNAs that are predicted to be translated within axon growth cones or dendrites have long half-lives while mRNAs encoding transcription factors that regulate neurogenesis have short half-lives. A search for candidate cis-regulatory elements identified enrichment of the Pumilio recognition element (PRE) in mRNAs encoding regulators of neurogenesis. We found that decreased expression of the RNA-binding protein Pumilio stabilized predicted neural mRNA targets and that a PRE is necessary to trigger reporter-transcript decay in the nervous system. Conclusions We found that differential mRNA decay contributes to the relative abundance of transcripts involved in cell-fate decisions, axonogenesis, and other critical events during Drosophila neural development. Neural-specific decay kinetics and the functional specificity of mRNA decay suggest the existence of a dynamic neurodevelopmental mRNA decay network. We found that Pumilio is one component of this network, revealing a novel function for this RNA-binding protein. Electronic supplementary material The online version of this article (doi:10.1186/s13064-015-0038-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dana A Burow
- Quantitative and Systems Biology Graduate Program, University of California, 5200 N. Lake Rd, Merced, CA, USA.
| | - Maxine C Umeh-Garcia
- Quantitative and Systems Biology Graduate Program, University of California, 5200 N. Lake Rd, Merced, CA, USA.
| | - Marie B True
- Quantitative and Systems Biology Graduate Program, University of California, 5200 N. Lake Rd, Merced, CA, USA.
| | - Crystal D Bakhaj
- Quantitative and Systems Biology Graduate Program, University of California, 5200 N. Lake Rd, Merced, CA, USA.
| | - David H Ardell
- Quantitative and Systems Biology Graduate Program, University of California, 5200 N. Lake Rd, Merced, CA, USA.
| | - Michael D Cleary
- Quantitative and Systems Biology Graduate Program, University of California, 5200 N. Lake Rd, Merced, CA, USA.
| |
Collapse
|
30
|
Abstract
Synaptic vesicles release their vesicular contents to the extracellular space by Ca(2+)-triggered exocytosis. The Ca(2+)-triggered exocytotic process is regulated by synaptotagmin (Syt), a vesicular Ca(2+)-binding C2 domain protein. Synaptotagmin 1 (Syt1), the most studied major isoform among 16 Syt isoforms, mediates Ca(2+)-triggered synaptic vesicle exocytosis by interacting with the target membranes and SNARE/complexin complex. In synapses of the central nervous system, synaptobrevin 2, a major vesicular SNARE protein, forms a ternary SNARE complex with the plasma membrane SNARE proteins, syntaxin 1 and SNAP25. The affinities of Ca(2+)-dependent interactions between Syt1 and its targets (i.e., SNARE complexes and membranes) are well correlated with the efficacies of the corresponding exocytotic processes. Therefore, different SNARE protein isoforms and membrane lipids, which interact with Syt1 with various affinities, are capable of regulating the efficacy of Syt1-mediated exocytosis. Otoferlin, another type of vesicular C2 domain protein that binds to the membrane in a Ca(2+)-dependent manner, is also involved in the Ca(2+)-triggered synaptic vesicle exocytosis in auditory hair cells. However, the functions of otoferlin in the exocytotic process are not well understood. In addition, at least five different types of synaptic vesicle proteins such as synaptic vesicle protein 2, cysteine string protein α, rab3, synapsin, and a group of proteins containing four transmembrane regions, which includes synaptophysin, synaptogyrin, and secretory carrier membrane protein, are involved in modulating the exocytotic process by regulating the formation and trafficking of synaptic vesicles.
Collapse
Affiliation(s)
- Ok-Ho Shin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
31
|
Xing G, Gan G, Chen D, Sun M, Yi J, Lv H, Han J, Xie W. Drosophila neuroligin3 regulates neuromuscular junction development and synaptic differentiation. J Biol Chem 2014; 289:31867-31877. [PMID: 25228693 DOI: 10.1074/jbc.m114.574897] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuroligins (Nlgs) are a family of cell adhesion molecules thought to be important for synapse maturation and function. Mammalian studies have shown that different Nlgs have different roles in synaptic maturation and function. In Drosophila melanogaster, the roles of Drosophila neuroligin1 (DNlg1), neuroligin2, and neuroligin4 have been examined. However, the roles of neuroligin3 (dnlg3) in synaptic development and function have not been determined. In this study, we used the Drosophila neuromuscular junctions (NMJs) as a model system to investigate the in vivo role of dnlg3. We showed that DNlg3 was expressed in both the CNS and NMJs where it was largely restricted to the postsynaptic site. We generated dnlg3 mutants and showed that these mutants exhibited an increased bouton number and reduced bouton size compared with the wild-type (WT) controls. Consistent with alterations in bouton properties, pre- and postsynaptic differentiations were affected in dnlg3 mutants. This included abnormal synaptic vesicle endocytosis, increased postsynaptic density length, and reduced GluRIIA recruitment. In addition to impaired synaptic development and differentiation, we found that synaptic transmission was reduced in dnlg3 mutants. Altogether, our data showed that DNlg3 was required for NMJ development, synaptic differentiation, and function.
Collapse
Affiliation(s)
- Guanglin Xing
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Guangming Gan
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Dandan Chen
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Mingkuan Sun
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Jukang Yi
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Huihui Lv
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Junhai Han
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Wei Xie
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China.
| |
Collapse
|
32
|
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: 1.4] [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
|
33
|
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: 1.6] [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
|
34
|
Abstract
Synaptic vesicle recycling is one of the best-studied cellular pathways. Many of the proteins involved are known, and their interactions are becoming increasingly clear. However, as for many other pathways, it is still difficult to understand synaptic vesicle recycling as a whole. While it is generally possible to point out how synaptic reactions take place, it is not always easy to understand what triggers or controls them. Also, it is often difficult to understand how the availability of the reaction partners is controlled: how the reaction partners manage to find each other in the right place, at the right time. I present here an overview of synaptic vesicle recycling, discussing the mechanisms that trigger different reactions, and those that ensure the availability of reaction partners. A central argument is that synaptic vesicles bind soluble cofactor proteins, with low affinity, and thus control their availability in the synapse, forming a buffer for cofactor proteins. The availability of cofactor proteins, in turn, regulates the different synaptic reactions. Similar mechanisms, in which one of the reaction partners buffers another, may apply to many other processes, from the biogenesis to the degradation of the synaptic vesicle.
Collapse
Affiliation(s)
- Silvio O Rizzoli
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen European Neuroscience Institute, Göttingen, Germany
| |
Collapse
|
35
|
Ye R, Carneiro AMD, Han Q, Airey D, Sanders-Bush E, Zhang B, Lu L, Williams R, Blakely RD. Quantitative trait loci mapping and gene network analysis implicate protocadherin-15 as a determinant of brain serotonin transporter expression. GENES BRAIN AND BEHAVIOR 2014; 13:261-75. [PMID: 24405699 DOI: 10.1111/gbb.12119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 12/15/2022]
Abstract
Presynaptic serotonin (5-hydroxytryptamine, 5-HT) transporters (SERT) regulate 5-HT signaling via antidepressant-sensitive clearance of released neurotransmitter. Polymorphisms in the human SERT gene (SLC6A4) have been linked to risk for multiple neuropsychiatric disorders, including depression, obsessive-compulsive disorder and autism. Using BXD recombinant inbred mice, a genetic reference population that can support the discovery of novel determinants of complex traits, merging collective trait assessments with bioinformatics approaches, we examine phenotypic and molecular networks associated with SERT gene and protein expression. Correlational analyses revealed a network of genes that significantly associated with SERT mRNA levels. We quantified SERT protein expression levels and identified region- and gender-specific quantitative trait loci (QTLs), one of which associated with male midbrain SERT protein expression, centered on the protocadherin-15 gene (Pcdh15), overlapped with a QTL for midbrain 5-HT levels. Pcdh15 was also the only QTL-associated gene whose midbrain mRNA expression significantly associated with both SERT protein and 5-HT traits, suggesting an unrecognized role of the cell adhesion protein in the development or function of 5-HT neurons. To test this hypothesis, we assessed SERT protein and 5-HT traits in the Pcdh15 functional null line (Pcdh15(av-) (3J) ), studies that revealed a strong, negative influence of Pcdh15 on these phenotypes. Together, our findings illustrate the power of multidimensional profiling of recombinant inbred lines in the analysis of molecular networks that support synaptic signaling, and that, as in the case of Pcdh15, can reveal novel relationships that may underlie risk for mental illness.
Collapse
Affiliation(s)
- R Ye
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Parkinson W, Dear ML, Rushton E, Broadie K. N-glycosylation requirements in neuromuscular synaptogenesis. Development 2013; 140:4970-81. [PMID: 24227656 DOI: 10.1242/dev.099192] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neural development requires N-glycosylation regulation of intercellular signaling, but the requirements in synaptogenesis have not been well tested. All complex and hybrid N-glycosylation requires MGAT1 (UDP-GlcNAc:α-3-D-mannoside-β1,2-N-acetylglucosaminyl-transferase I) function, and Mgat1 nulls are the most compromised N-glycosylation condition that survive long enough to permit synaptogenesis studies. At the Drosophila neuromuscular junction (NMJ), Mgat1 mutants display selective loss of lectin-defined carbohydrates in the extracellular synaptomatrix, and an accompanying accumulation of the secreted endogenous Mind the gap (MTG) lectin, a key synaptogenesis regulator. Null Mgat1 mutants exhibit strongly overelaborated synaptic structural development, consistent with inhibitory roles for complex/hybrid N-glycans in morphological synaptogenesis, and strengthened functional synapse differentiation, consistent with synaptogenic MTG functions. Synapse molecular composition is surprisingly selectively altered, with decreases in presynaptic active zone Bruchpilot (BRP) and postsynaptic Glutamate receptor subtype B (GLURIIB), but no detectable change in a wide range of other synaptic components. Synaptogenesis is driven by bidirectional trans-synaptic signals that traverse the glycan-rich synaptomatrix, and Mgat1 mutation disrupts both anterograde and retrograde signals, consistent with MTG regulation of trans-synaptic signaling. Downstream of intercellular signaling, pre- and postsynaptic scaffolds are recruited to drive synaptogenesis, and Mgat1 mutants exhibit loss of both classic Discs large 1 (DLG1) and newly defined Lethal (2) giant larvae [L(2)GL] scaffolds. We conclude that MGAT1-dependent N-glycosylation shapes the synaptomatrix carbohydrate environment and endogenous lectin localization within this domain, to modulate retention of trans-synaptic signaling ligands driving synaptic scaffold recruitment during synaptogenesis.
Collapse
Affiliation(s)
- William Parkinson
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37212, USA
| | | | | | | |
Collapse
|
37
|
Poudel KR, Bai J. Synaptic vesicle morphology: a case of protein sorting? Curr Opin Cell Biol 2013; 26:28-33. [PMID: 24529243 DOI: 10.1016/j.ceb.2013.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 10/26/2022]
Abstract
Synaptic vesicles (SVs) are the repositories of neurotransmitters. They are locally recycled at nerve terminals following exocytosis. A unique feature of these vesicles is the uniformity of their morphology, which is well maintained even after rounds of exocytosis and endocytosis. Several studies suggest that disruption of clathrin adaptor proteins leads to defects in sorting cargoes and alterations in SV morphology. Here, we review the links between adaptor proteins and SV size, and highlight how protein sorting may impact SV architecture. Molecular players such as clathrin, adaptor proteins, accessory proteins, SV cargoes and lipid composition may act together to establish a stable regulatory network to maintain SV morphology.
Collapse
Affiliation(s)
- Kumud R Poudel
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Jihong Bai
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Biochemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|