1
|
Wang H, Hoffmann C, Tromm JV, Su X, Elliott J, Wang H, Baum J, Pang ZP, Milovanovic D, Shi Z. Live-Cell Quantification Reveals Viscoelastic Regulation of Synapsin Condensates by α-Synuclein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605529. [PMID: 39211102 PMCID: PMC11361170 DOI: 10.1101/2024.07.28.605529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Synapsin and α-synuclein represent a growing list of condensate-forming proteins where the material states of condensates are directly linked to cellular functions (e.g., neurotransmission) and pathology (e.g., neurodegeneration). However, quantifying condensate material properties in living systems has been a significant challenge. To address this, we develop MAPAC (micropipette aspiration and whole-cell patch clamp), a platform that allows direct material quantification of condensates in live cells. We find 10,000-fold variations in the viscoelasticity of synapsin condensates, regulated by the partitioning of α-synuclein, a marker for synucleinopathies. Through in vitro reconstitutions, we identify 4 molecular factors that distinctly regulate the viscosity and interfacial tension of synapsin condensates, verifying the cellular effects of α-synuclein. Overall, our study provides unprecedented quantitative insights into the material properties of neuronal condensates and reveals a crucial role of α-synuclein in regulating condensate viscoelasticity. Furthermore, we envision MAPAC applicable to study a broad range of condensates in vivo. .
Collapse
|
2
|
Bruentgens F, Moreno Velasquez L, Stumpf A, Parthier D, Breustedt J, Benfenati F, Milovanovic D, Schmitz D, Orlando M. The Lack of Synapsin Alters Presynaptic Plasticity at Hippocampal Mossy Fibers in Male Mice. eNeuro 2024; 11:ENEURO.0330-23.2024. [PMID: 38866497 PMCID: PMC11223178 DOI: 10.1523/eneuro.0330-23.2024] [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: 08/29/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Synapsins are highly abundant presynaptic proteins that play a crucial role in neurotransmission and plasticity via the clustering of synaptic vesicles. The synapsin III isoform is usually downregulated after development, but in hippocampal mossy fiber boutons, it persists in adulthood. Mossy fiber boutons express presynaptic forms of short- and long-term plasticity, which are thought to underlie different forms of learning. Previous research on synapsins at this synapse focused on synapsin isoforms I and II. Thus, a complete picture regarding the role of synapsins in mossy fiber plasticity is still missing. Here, we investigated presynaptic plasticity at hippocampal mossy fiber boutons by combining electrophysiological field recordings and transmission electron microscopy in a mouse model lacking all synapsin isoforms. We found decreased short-term plasticity, i.e., decreased facilitation and post-tetanic potentiation, but increased long-term potentiation in male synapsin triple knock-out (KO) mice. At the ultrastructural level, we observed more dispersed vesicles and a higher density of active zones in mossy fiber boutons from KO animals. Our results indicate that all synapsin isoforms are required for fine regulation of short- and long-term presynaptic plasticity at the mossy fiber synapse.
Collapse
Affiliation(s)
- Felicitas Bruentgens
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Laura Moreno Velasquez
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Alexander Stumpf
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Daniel Parthier
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Jörg Breustedt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa 16163, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Dragomir Milovanovic
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Einstein Center for Neurosciences, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin 10117, Germany
| | - Dietmar Schmitz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Einstein Center for Neurosciences, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Berlin 10115, Germany
| | - Marta Orlando
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| |
Collapse
|
3
|
Cuhadar U, Calzado-Reyes L, Pascual-Caro C, Aberra AS, Ritzau-Jost A, Aggarwal A, Ibata K, Podgorski K, Yuzaki M, Geis C, Hallerman S, Hoppa MB, de Juan-Sanz J. Activity-driven synaptic translocation of LGI1 controls excitatory neurotransmission. Cell Rep 2024; 43:114186. [PMID: 38700985 PMCID: PMC11156761 DOI: 10.1016/j.celrep.2024.114186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 12/14/2023] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
The fine control of synaptic function requires robust trans-synaptic molecular interactions. However, it remains poorly understood how trans-synaptic bridges change to reflect the functional states of the synapse. Here, we develop optical tools to visualize in firing synapses the molecular behavior of two trans-synaptic proteins, LGI1 and ADAM23, and find that neuronal activity acutely rearranges their abundance at the synaptic cleft. Surprisingly, synaptic LGI1 is primarily not secreted, as described elsewhere, but exo- and endocytosed through its interaction with ADAM23. Activity-driven translocation of LGI1 facilitates the formation of trans-synaptic connections proportionally to the history of activity of the synapse, adjusting excitatory transmission to synaptic firing rates. Accordingly, we find that patient-derived autoantibodies against LGI1 reduce its surface fraction and cause increased glutamate release. Our findings suggest that LGI1 abundance at the synaptic cleft can be acutely remodeled and serves as a critical control point for synaptic function.
Collapse
Affiliation(s)
- Ulku Cuhadar
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| | - Lorenzo Calzado-Reyes
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| | - Carlos Pascual-Caro
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| | - Aman S Aberra
- Department of Biology, Dartmouth College, Hanover, NH 03755, USA
| | - Andreas Ritzau-Jost
- Carl-Ludwig-Institute of Physiology, Faculty of Medicine, Leipzig University, 04317 Leipzig, Germany
| | - Abhi Aggarwal
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Keiji Ibata
- Department of Neurophysiology, Keio University, Tokyo 160-8582, Japan
| | | | - Michisuke Yuzaki
- Department of Neurophysiology, Keio University, Tokyo 160-8582, Japan
| | - Christian Geis
- Department of Neurology, Section Translational Neuroimmunology, Jena University Hospital, 07747 Jena, Germany
| | - Stefan Hallerman
- Carl-Ludwig-Institute of Physiology, Faculty of Medicine, Leipzig University, 04317 Leipzig, Germany
| | - Michael B Hoppa
- Department of Biology, Dartmouth College, Hanover, NH 03755, USA
| | - Jaime de Juan-Sanz
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France.
| |
Collapse
|
4
|
Le Dréan ME, Le Berre-Scoul C, Paillé V, Caillaud M, Oullier T, Gonzales J, Hulin P, Neunlist M, Talon S, Boudin H. The regulation of enteric neuron connectivity by semaphorin 5A is affected by the autism-associated S956G missense mutation. iScience 2024; 27:109638. [PMID: 38650986 PMCID: PMC11033180 DOI: 10.1016/j.isci.2024.109638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
The neural network of the enteric nervous system (ENS) underlies gastrointestinal functions. However, the molecular mechanisms involved in enteric neuronal connectivity are poorly characterized. Here, we studied the role of semaphorin 5A (Sema5A), previously characterized in the central nervous system, on ENS neuronal connectivity. Sema5A is linked to autism spectrum disorder (ASD), a neurodevelopmental disorder frequently associated with gastrointestinal comorbidities, and potentially associated with ENS impairments. This study investigated in rat enteric neuron cultures and gut explants the role of Sema5A on enteric neuron connectivity and the impact of ASD-associated mutations on Sema5A activity. Our findings demonstrated that Sema5A promoted axonal complexity and reduced functional connectivity in enteric neurons. Strikingly, the ASD-associated mutation S956G in Sema5A strongly affected these activities. This study identifies a critical role of Sema5A in the ENS as a regulator of neuronal connectivity that might be compromised in ASD.
Collapse
Affiliation(s)
- Morgane E. Le Dréan
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Catherine Le Berre-Scoul
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Vincent Paillé
- Nantes Université, INRAE, UMR 1280, PhAN, IMAD, 44000 Nantes, France
| | - Martial Caillaud
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Thibauld Oullier
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Jacques Gonzales
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Philippe Hulin
- Plateforme MicroPICell Nantes Université, CHU Nantes, CNRS, INSERM, BioCore, US16, SFR Bonamy, Nantes, France
| | - Michel Neunlist
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Sophie Talon
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Hélène Boudin
- Nantes Université, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| |
Collapse
|
5
|
Rodriguez Gotor JJ, Mahfooz K, Perez-Otano I, Wesseling JF. Parallel processing of quickly and slowly mobilized reserve vesicles in hippocampal synapses. eLife 2024; 12:RP88212. [PMID: 38727712 PMCID: PMC11087054 DOI: 10.7554/elife.88212] [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] [Indexed: 05/12/2024] Open
Abstract
Vesicles within presynaptic terminals are thought to be segregated into a variety of readily releasable and reserve pools. The nature of the pools and trafficking between them is not well understood, but pools that are slow to mobilize when synapses are active are often assumed to feed pools that are mobilized more quickly, in a series. However, electrophysiological studies of synaptic transmission have suggested instead a parallel organization where vesicles within slowly and quickly mobilized reserve pools would separately feed independent reluctant- and fast-releasing subdivisions of the readily releasable pool. Here, we use FM-dyes to confirm the existence of multiple reserve pools at hippocampal synapses and a parallel organization that prevents intermixing between the pools, even when stimulation is intense enough to drive exocytosis at the maximum rate. The experiments additionally demonstrate extensive heterogeneity among synapses in the relative sizes of the slowly and quickly mobilized reserve pools, which suggests equivalent heterogeneity in the numbers of reluctant and fast-releasing readily releasable vesicles that may be relevant for understanding information processing and storage.
Collapse
Affiliation(s)
| | - Kashif Mahfooz
- Department of Pharmacology, University of OxfordOxfordUnited Kingdom
| | - Isabel Perez-Otano
- Instituto de Neurociencias de Alicante CSIC-UMHSan Juan de AlicanteSpain
| | - John F Wesseling
- Instituto de Neurociencias de Alicante CSIC-UMHSan Juan de AlicanteSpain
| |
Collapse
|
6
|
Farrell RJ, Bredvik KG, Hoppa MB, Hennigan ST, Brown TA, Ryan TA. A ratiometric ER calcium sensor for quantitative comparisons across cell types and subcellular regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580492. [PMID: 38405980 PMCID: PMC10888930 DOI: 10.1101/2024.02.15.580492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The endoplasmic reticulum (ER) is an important regulator of Ca 2 + in cells and dysregulation of ER calcium homeostasis can lead to numerous pathologies. Understanding how various pharmacological and genetic perturbations of ER Ca 2 + homeostasis impacts cellular physiology would likely be facilitated by more quantitative measurements of ER Ca 2 + levels that allow easier comparisons across conditions. Here, we developed a ratiometric version of our original ER-GCaMP probe that allows for more quantitative comparisons of the concentration of Ca 2 + in the ER across cell types and sub-cellular compartments. Using this approach we show that the resting concentration of ER Ca2+ in primary dissociated neurons is substantially lower than that in measured in embryonic fibroblasts.
Collapse
Affiliation(s)
- Ryan J Farrell
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- David Rockefeller Graduate Program, The Rockefeller University, New York, NY, USA, 10065
- Present Address: Neuroscience Institute, NYU Medical Center, New York, NY, USA, 10016
| | - Kirsten G Bredvik
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Tri-Institutional M.D./Ph.D. Program, Weill Cornell Medicine, New York, NY, USA, 10065
| | - Michael B Hoppa
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Present Address: Department of Biology, Dartmouth College, Hanover, NH 03755
| | - S Thomas Hennigan
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147
| | - Timothy A Brown
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Lead Contact: correspondence
| |
Collapse
|
7
|
Mao LM, Thallapureddy K, Wang JQ. Effects of propofol on presynaptic synapsin phosphorylation in the mouse brain in vivo. Brain Res 2024; 1823:148671. [PMID: 37952872 PMCID: PMC10806815 DOI: 10.1016/j.brainres.2023.148671] [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: 08/10/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The commonly used general anesthetic propofol can enhance the γ-aminobutyric acid-mediated inhibitory synaptic transmission and depress the glutamatergic excitatory synaptic transmission to achieve general anesthesia and other outcomes. In addition to the actions at postsynaptic sites, the modulation of presynaptic activity by propofol is thought to contribute to neurophysiological effects of the anesthetic, although potential targets of propofol within presynaptic nerve terminals are incompletely studied at present. In this study, we explored the possible linkage of propofol to synapsins, a family of neuron-specific phosphoproteins which are the most abundant proteins on presynaptic vesicles, in the adult mouse brain in vivo. We found that an intraperitoneal injection of propofol at a dose that caused loss of righting reflex increased basal levels of synapsin phosphorylation at the major representative phosphorylation sites (serine 9, serine 62/67, and serine 603) in the prefrontal cortex (PFC) of male and female mice. Propofol also elevated synapsin phosphorylation at these sites in the striatum and S9 and S62/67 phosphorylation in the hippocampus, while propofol had no effect on tyrosine hydroxylase phosphorylation in striatal nerve terminals. Total synapsin protein expression in the PFC, hippocampus, and striatum was not altered by propofol. These results reveal that synapsin could be a novel substrate of propofol in the presynaptic neurotransmitter release machinery. Propofol possesses the ability to upregulate synapsin phosphorylation in broad mouse brain regions.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Khyathi Thallapureddy
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
8
|
Moors TE, Milovanovic D. Defining a Lewy Body: Running Up the Hill of Shifting Definitions and Evolving Concepts. JOURNAL OF PARKINSON'S DISEASE 2024; 14:17-33. [PMID: 38189713 PMCID: PMC10836569 DOI: 10.3233/jpd-230183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/14/2023] [Indexed: 01/09/2024]
Abstract
Lewy bodies (LBs) are pathological hallmarks of Parkinson's disease and dementia with Lewy bodies, characterized by the accumulation of α-synuclein (αSyn) protein in the brain. While LBs were first described a century ago, their formation and morphogenesis mechanisms remain incompletely understood. Here, we present a historical overview of LB definitions and highlight the importance of semantic clarity and precise definitions when describing brain inclusions. Recent breakthroughs in imaging revealed shared features within LB subsets and the enrichment of membrane-bound organelles in these structures, challenging the conventional LB formation model. We discuss the involvement of emerging concepts of liquid-liquid phase separation, where biomolecules demix from a solution to form dense condensates, as a potential LB formation mechanism. Finally, we emphasize the need for the operational definitions of LBs based on morphological characteristics and detection protocols, particularly in studies investigating LB formation mechanisms. A better understanding of LB organization and ultrastructure can contribute to the development of targeted therapeutic strategies for synucleinopathies.
Collapse
Affiliation(s)
- Tim E. Moors
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Einstein Center for Neuroscience, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
9
|
Hersey M, Tanda G. Modafinil, an atypical CNS stimulant? ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:287-326. [PMID: 38467484 DOI: 10.1016/bs.apha.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Modafinil is a central nervous system stimulant approved for the treatment of narcolepsy and sleep disorders. Due to its wide range of biochemical actions, modafinil has been explored for other potential therapeutic uses. Indeed, it has shown promise as a therapy for cognitive disfunction resulting from neurologic disorders like ADHD, and as a smart drug in non-medical settings. The mechanism(s) of actions underlying the therapeutic efficacy of this agent remains largely elusive. Modafinil is known to inhibit the dopamine transporter, thus decreasing dopamine reuptake following neuronal release, an effect shared by addictive psychostimulants. However, modafinil is unique in that only a few cases of dependence on this drug have been reported, as compared to other psychostimulants. Moreover, modafinil has been tested, with some success, as a potential therapeutic agent to combat psychostimulant and other substance use disorders. Modafinil has additional, but less understood, actions on other neurotransmitter systems (GABA, glutamate, serotonin, norepinephrine, etc.). These interactions, together with its ability to activate selected brain regions, are likely one of the keys to understand its unique pharmacology and therapeutic activity as a CNS stimulant. In this chapter, we outline the pharmacokinetics and pharmacodynamics of modafinil that suggest it has an "atypical" CNS stimulant profile. We also highlight the current approved and off label uses of modafinil, including its beneficial effects as a treatment for sleep disorders, cognitive functions, and substance use disorders.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, NIDA-IRP, NIH, Baltimore, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, NIDA-IRP, NIH, Baltimore, MD, United States.
| |
Collapse
|
10
|
Li Q, Song Q, Guo W, Cao Y, Cui X, Chen D, Shum HC. Synthetic Membraneless Droplets for Synaptic-Like Clustering of Lipid Vesicles. Angew Chem Int Ed Engl 2023; 62:e202313096. [PMID: 37728515 DOI: 10.1002/anie.202313096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/21/2023]
Abstract
In eukaryotic cells, the membraneless organelles (MLOs) formed via liquid-liquid phase separation (LLPS) are found to interact intimately with membranous organelles (MOs). One major mode is the clustering of MOs by MLOs, such as the formation of clusters of synaptic vesicles at nerve terminals mediated by the synapsin-rich MLOs. Aqueous droplets, including complex coacervates and aqueous two-phase systems, have been plausible MLO-mimics to emulate or elucidate biological processes. However, neither of them can cluster lipid vesicles (LVs) like MLOs. In this work, we develop a synthetic droplet assembled from a combination of two different interactions underlying the formation of these two droplets, namely, associative and segregative interactions, which we call segregative-associative (SA) droplets. The SA droplets cluster and disperse LVs recapitulating the key functional features of synapsin condensates, which can be attributed to the weak electrostatic interaction environment provided by SA droplets. This work suggests LLPS with combined segregative and associative interactions as a possible route for synaptic clustering of lipid vesicles and highlights SA droplets as plausible MLO-mimics and models for studying and mimicking related cellular dynamics.
Collapse
Affiliation(s)
- Qingchuan Li
- School of Chemistry & Chemical Engineering, National Engineering Research Center for Colloidal Materials, Shandong University, 27 Shanda Nanlu, Jinan, Shandong, P.R.China
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR), Hong Kong, China
| | - Qingchun Song
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Wei Guo
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR), Hong Kong, China
| | - Yang Cao
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xinyu Cui
- Department of Public Health, Mudanjiang Medical University, Mudanjiang, 157000, P. R. China
| | - Dairong Chen
- School of Chemistry & Chemical Engineering, National Engineering Research Center for Colloidal Materials, Shandong University, 27 Shanda Nanlu, Jinan, Shandong, P.R.China
| | - Ho Cheung Shum
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR), Hong Kong, China
| |
Collapse
|
11
|
Xie RG, Xu GY, Wu SX, Luo C. Presynaptic glutamate receptors in nociception. Pharmacol Ther 2023; 251:108539. [PMID: 37783347 DOI: 10.1016/j.pharmthera.2023.108539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Chronic pain is a frequent, distressing and poorly understood health problem. Plasticity of synaptic transmission in the nociceptive pathways after inflammation or injury is assumed to be an important cellular basis for chronic, pathological pain. Glutamate serves as the main excitatory neurotransmitter at key synapses in the somatosensory nociceptive pathways, in which it acts on both ionotropic and metabotropic glutamate receptors. Although conventionally postsynaptic, compelling anatomical and physiological evidence demonstrates the presence of presynaptic glutamate receptors in the nociceptive pathways. Presynaptic glutamate receptors play crucial roles in nociceptive synaptic transmission and plasticity. They modulate presynaptic neurotransmitter release and synaptic plasticity, which in turn regulates pain sensitization. In this review, we summarize the latest understanding of the expression of presynaptic glutamate receptors in the nociceptive pathways, and how they contribute to nociceptive information processing and pain hypersensitivity associated with inflammation / injury. We uncover the cellular and molecular mechanisms of presynaptic glutamate receptors in shaping synaptic transmission and plasticity to mediate pain chronicity, which may provide therapeutic approaches for treatment of chronic pain.
Collapse
Affiliation(s)
- Rou-Gang Xie
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Sheng-Xi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
12
|
Lim CM, González Díaz A, Fuxreiter M, Pun FW, Zhavoronkov A, Vendruscolo M. Multiomic prediction of therapeutic targets for human diseases associated with protein phase separation. Proc Natl Acad Sci U S A 2023; 120:e2300215120. [PMID: 37774095 PMCID: PMC10556643 DOI: 10.1073/pnas.2300215120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/02/2023] [Indexed: 10/01/2023] Open
Abstract
The phenomenon of protein phase separation (PPS) underlies a wide range of cellular functions. Correspondingly, the dysregulation of the PPS process has been associated with numerous human diseases. To enable therapeutic interventions based on the regulation of this association, possible targets should be identified. For this purpose, we present an approach that combines the multiomic PandaOmics platform with the FuzDrop method to identify PPS-prone disease-associated proteins. Using this approach, we prioritize candidates with high PandaOmics and FuzDrop scores using a profiling method that accounts for a wide range of parameters relevant for disease mechanism and pharmacological intervention. We validate the differential phase separation behaviors of three predicted Alzheimer's disease targets (MARCKS, CAMKK2, and p62) in two cell models of this disease. Overall, the approach that we present generates a list of possible therapeutic targets for human diseases associated with the dysregulation of the PPS process.
Collapse
Affiliation(s)
- Christine M. Lim
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, CambridgeCB2 1EW, United Kingdom
| | - Alicia González Díaz
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, CambridgeCB2 1EW, United Kingdom
| | - Monika Fuxreiter
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
| | - Frank W. Pun
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong, China
| | - Alex Zhavoronkov
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong, China
| | - Michele Vendruscolo
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, CambridgeCB2 1EW, United Kingdom
| |
Collapse
|
13
|
Xu H, Oses-Prieto JA, Khvotchev M, Jain S, Liang J, Burlingame A, Edwards RH. Adaptor protein AP-3 produces synaptic vesicles that release at high frequency by recruiting phospholipid flippase ATP8A1. Nat Neurosci 2023; 26:1685-1700. [PMID: 37723322 DOI: 10.1038/s41593-023-01434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023]
Abstract
Neural systems encode information in the frequency of action potentials, which is then decoded by synaptic transmission. However, the rapid, synchronous release of neurotransmitters depletes synaptic vesicles (SVs), limiting release at high firing rates. How then do synapses convey information about frequency? Here, we show in mouse hippocampal neurons and slices that the adaptor protein AP-3 makes a subset of SVs that respond specifically to high-frequency stimulation. Neurotransmitter transporters slot onto these SVs in different proportions, contributing to the distinct properties of release observed at different excitatory synapses. Proteomics reveals that AP-3 targets the phospholipid flippase ATP8A1 to SVs; loss of ATP8A1 recapitulates the defect in SV mobilization at high frequency observed with loss of AP-3. The mechanism involves recruitment of synapsin by the cytoplasmically oriented phosphatidylserine translocated by ATP8A1. Thus, ATP8A1 enables the subset of SVs made by AP-3 to release at high frequency.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Mikhail Khvotchev
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Shweta Jain
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Jocelyn Liang
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Alma Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Robert H Edwards
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
14
|
Song SH, Augustine GJ. Different mechanisms of synapsin-induced vesicle clustering at inhibitory and excitatory synapses. Cell Rep 2023; 42:113004. [PMID: 37597184 DOI: 10.1016/j.celrep.2023.113004] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/26/2023] [Accepted: 08/02/2023] [Indexed: 08/21/2023] Open
Abstract
Synapsins cluster synaptic vesicles (SVs) to provide a reserve pool (RP) of SVs that maintains synaptic transmission during sustained activity. However, it is unclear how synapsins cluster SVs. Here we show that either liquid-liquid phase separation (LLPS) or tetramerization-dependent cross-linking can cluster SVs, depending on whether a synapse is excitatory or inhibitory. Cell-free reconstitution reveals that both mechanisms can cluster SVs, with tetramerization being more effective. At inhibitory synapses, perturbing synapsin-dependent LLPS impairs SV clustering and synchronization of gamma-aminobutyric acid (GABA) release, while preventing synapsin tetramerization does not. At glutamatergic synapses, the opposite is true: synapsin tetramerization enhances clustering of glutamatergic SVs and mobilization of these SVs from the RP, while synapsin LLPS does not. Comparison of inhibitory and excitatory transmission during prolonged synaptic activity reveals that synapsin LLPS serves as a brake to limit GABA release, while synapsin tetramerization enables rapid mobilization of SVs from the RP to sustain glutamate release.
Collapse
Affiliation(s)
- Sang-Ho Song
- Neuroscience and Mental Health Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - George J Augustine
- Neuroscience and Mental Health Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|
15
|
Yoshida T, Takenaka KI, Sakamoto H, Kojima Y, Sakano T, Shibayama K, Nakamura K, Hanawa-Suetsugu K, Mori Y, Hirabayashi Y, Hirose K, Takamori S. Compartmentalization of soluble endocytic proteins in synaptic vesicle clusters by phase separation. iScience 2023; 26:106826. [PMID: 37250768 PMCID: PMC10209458 DOI: 10.1016/j.isci.2023.106826] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/10/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Synaptic vesicle (SV) clusters, which reportedly result from synapsin's capacity to undergo liquid-liquid phase separation (LLPS), constitute the structural basis for neurotransmission. Although these clusters contain various endocytic accessory proteins, how endocytic proteins accumulate in SV clusters remains unknown. Here, we report that endophilin A1 (EndoA1), the endocytic scaffold protein, undergoes LLPS under physiologically relevant concentrations at presynaptic terminals. On heterologous expression, EndoA1 facilitates the formation of synapsin condensates and accumulates in SV-like vesicle clusters via synapsin. Moreover, EndoA1 condensates recruit endocytic proteins such as dynamin 1, amphiphysin, and intersectin 1, none of which are recruited in vesicle clusters by synapsin. In cultured neurons, like synapsin, EndoA1 is compartmentalized in SV clusters through LLPS, exhibiting activity-dependent dispersion/reassembly cycles. Thus, beyond its essential function in SV endocytosis, EndoA1 serves an additional structural function by undergoing LLPS, thereby accumulating various endocytic proteins in dynamic SV clusters in concert with synapsin.
Collapse
Affiliation(s)
- Tomofumi Yoshida
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Koh-ichiro Takenaka
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Hirokazu Sakamoto
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yusuke Kojima
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Takumi Sakano
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-0033, Japan
| | - Koyo Shibayama
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-0033, Japan
| | - Koki Nakamura
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kyoko Hanawa-Suetsugu
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Yasunori Mori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Yusuke Hirabayashi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| |
Collapse
|
16
|
Keighron JD, Bonaventura J, Li Y, Yang JW, DeMarco EM, Hersey M, Cao J, Sandtner W, Michaelides M, Sitte HH, Newman AH, Tanda G. Interactions of calmodulin kinase II with the dopamine transporter facilitate cocaine-induced enhancement of evoked dopamine release. Transl Psychiatry 2023; 13:202. [PMID: 37311803 PMCID: PMC10264427 DOI: 10.1038/s41398-023-02493-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/17/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023] Open
Abstract
Typical and atypical dopamine uptake inhibitors (DUIs) prefer distinct conformations of the dopamine transporter (DAT) to form ligand-transporter complexes, resulting in markedly different effects on behavior, neurochemistry, and potential for addiction. Here we show that cocaine and cocaine-like typical psychostimulants elicit changes in DA dynamics distinct from those elicited by atypical DUIs, as measured via voltammetry procedures. While both classes of DUIs reduced DA clearance rate, an effect significantly related to their DAT affinity, only typical DUIs elicited a significant stimulation of evoked DA release, an effect unrelated to their DAT affinity, which suggests a mechanism of action other than or in addition to DAT blockade. When given in combination, typical DUIs enhance the stimulatory effects of cocaine on evoked DA release while atypical DUIs blunt them. Pretreatments with an inhibitor of CaMKIIα, a kinase that interacts with DAT and that regulates synapsin phosphorylation and mobilization of reserve pools of DA vesicles, blunted the effects of cocaine on evoked DA release. Our results suggest a role for CaMKIIα in modulating the effects of cocaine on evoked DA release without affecting cocaine inhibition of DA reuptake. This effect is related to a specific DAT conformation stabilized by cocaine. Moreover, atypical DUIs, which prefer a distinct DAT conformation, blunt cocaine's neurochemical and behavioral effects, indicating a unique mechanism underlying their potential as medications for treating psychostimulant use disorder.
Collapse
Affiliation(s)
- Jacqueline D Keighron
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Department of Biological and Chemical Science, New York Institute of Technology, Old Westbury, NY, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Department of Pathology and Experimental Therapeutics, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain
| | - Yang Li
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Emily M DeMarco
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Melinda Hersey
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Walter Sandtner
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amy Hauck Newman
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Gianluigi Tanda
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.
| |
Collapse
|
17
|
Radecke J, Seeger R, Kádková A, Laugks U, Khosrozadeh A, Goldie KN, Lučić V, Sørensen JB, Zuber B. Morphofunctional changes at the active zone during synaptic vesicle exocytosis. EMBO Rep 2023; 24:e55719. [PMID: 36876590 PMCID: PMC10157379 DOI: 10.15252/embr.202255719] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/30/2023] [Accepted: 02/16/2023] [Indexed: 03/07/2023] Open
Abstract
Synaptic vesicle (SV) fusion with the plasma membrane (PM) proceeds through intermediate steps that remain poorly resolved. The effect of persistent high or low exocytosis activity on intermediate steps remains unknown. Using spray-mixing plunge-freezing cryo-electron tomography we observe events following synaptic stimulation at nanometer resolution in near-native samples. Our data suggest that during the stage that immediately follows stimulation, termed early fusion, PM and SV membrane curvature changes to establish a point contact. The next stage-late fusion-shows fusion pore opening and SV collapse. During early fusion, proximal tethered SVs form additional tethers with the PM and increase the inter-SV connector number. In the late-fusion stage, PM-proximal SVs lose their interconnections, allowing them to move toward the PM. Two SNAP-25 mutations, one arresting and one disinhibiting spontaneous release, cause connector loss. The disinhibiting mutation causes loss of membrane-proximal multiple-tethered SVs. Overall, tether formation and connector dissolution are triggered by stimulation and respond to spontaneous fusion rate manipulation. These morphological observations likely correspond to SV transition from one functional pool to another.
Collapse
Affiliation(s)
- Julika Radecke
- Institute of AnatomyUniversity of BernBernSwitzerland
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
- Diamond Light Source LtdDidcotUK
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Raphaela Seeger
- Institute of AnatomyUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Anna Kádková
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Ulrike Laugks
- Max‐Planck‐Institute of BiochemistryMartinsriedGermany
| | - Amin Khosrozadeh
- Institute of AnatomyUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | | | - Vladan Lučić
- Max‐Planck‐Institute of BiochemistryMartinsriedGermany
| | - Jakob B Sørensen
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Benoît Zuber
- Institute of AnatomyUniversity of BernBernSwitzerland
| |
Collapse
|
18
|
Cook DC, Ryan TA. GABA BR silencing of nerve terminals. eLife 2023; 12:e83530. [PMID: 37014052 PMCID: PMC10115440 DOI: 10.7554/elife.83530] [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: 09/17/2022] [Accepted: 04/03/2023] [Indexed: 04/05/2023] Open
Abstract
Control of neurotransmission efficacy is central to theories of how the brain computes and stores information. Presynaptic G-protein coupled receptors (GPCRs) are critical in this problem as they locally influence synaptic strength and can operate on a wide range of time scales. Among the mechanisms by which GPCRs impact neurotransmission is by inhibiting voltage-gated calcium (Ca2+) influx in the active zone. Here, using quantitative analysis of both single bouton Ca2+ influx and exocytosis, we uncovered an unexpected non-linear relationship between the magnitude of action potential driven Ca2+ influx and the concentration of external Ca2+ ([Ca2+]e). We find that this unexpected relationship is leveraged by GPCR signaling when operating at the nominal physiological set point for [Ca2+]e, 1.2 mM, to achieve complete silencing of nerve terminals. These data imply that the information throughput in neural circuits can be readily modulated in an all-or-none fashion at the single synapse level when operating at the physiological set point.
Collapse
Affiliation(s)
- Daniel C Cook
- Department of Anesthesiology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Timothy A Ryan
- Department of Anesthesiology, Weill Cornell Medical CollegeNew YorkUnited States
- Department of Biochemistry, Weill Cornell Medical CollegeNew YorkUnited States
| |
Collapse
|
19
|
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: 36] [Impact Index Per Article: 36.0] [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
|
20
|
Garner CC, Ackermann F. Synaptic logistics: The presynaptic scaffold protein Piccolo a nodal point tuning synaptic vesicle recycling, maintenance and integrity. Mol Cell Neurosci 2023; 124:103795. [PMID: 36436725 DOI: 10.1016/j.mcn.2022.103795] [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: 09/30/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Properly working synapses are one important guarantor for a functional and healthy brain. They are small, densely packed structures, where information is transmitted through the release of neurotransmitters from synaptic vesicles (SVs). The latter cycle within the presynaptic terminal as they first fuse with the plasma membrane to deliver their neurotransmitter, and afterwards become recycled and prepared for a new release event. The synapse is an autonomous structure functioning mostly independent of the neuronal soma. Dysfunction in synaptic processes associated with local insults or genetic abnormalities can directly compromise synapse function and integrity and subsequently lead to the onset of neurodegenerative diseases. Therefore, measures need to be in place counteracting these threats for instance through the continuous replacement of old and damaged SV proteins. Interestingly recent studies show that the presynaptic scaffolding protein Piccolo contributes to health, function and integrity of synapses, as it mediates the delivery of synaptic proteins from the trans-Golgi network (TGN) towards synapses, as well as the local recycling and turnover of SV proteins within synaptic terminals. It can fulfill these various tasks through its multi-domain structure and ability to interact with numerous binding partners. In addition, Piccolo has recently been linked with the early onset neurodegenerative disease Pontocerebellar Hypoplasia Type 3 (PCH3) further underlying its importance for neuronal health. In this review, we will focus on Piccolo's contributions to synapse function, health and integrity and make a connection how those may contribute to the disease pattern of PCH3.
Collapse
Affiliation(s)
- Craig C Garner
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Germany
| | - Frauke Ackermann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.
| |
Collapse
|
21
|
Sharma M, Burré J. α-Synuclein in synaptic function and dysfunction. Trends Neurosci 2023; 46:153-166. [PMID: 36567199 PMCID: PMC9877183 DOI: 10.1016/j.tins.2022.11.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
α-Synuclein is a neuronal protein that is enriched in presynaptic terminals. Under physiological conditions, it binds to synaptic vesicle membranes and functions in neurotransmitter release, although the molecular details remain unclear, and it is controversial whether α-synuclein inhibits or facilitates neurotransmitter release. Pathologically, in synucleinopathies including Parkinson's disease (PD), α-synuclein forms aggregates that recruit monomeric α-synuclein and spread throughout the brain, which triggers neuronal dysfunction at molecular, cellular, and organ levels. Here, we present an overview of the effects of α-synuclein on SNARE-complex assembly, neurotransmitter release, and synaptic vesicle pool homeostasis, and discuss how the observed divergent effects of α-synuclein on neurotransmitter release can be reconciled. We also discuss how gain-of-function versus loss-of-function of α-synuclein may contribute to pathogenesis in synucleinopathies.
Collapse
Affiliation(s)
- Manu Sharma
- Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Jacqueline Burré
- Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
22
|
Wu X, Qiu H, Zhang M. Interactions between Membraneless Condensates and Membranous Organelles at the Presynapse: A Phase Separation View of Synaptic Vesicle Cycle. J Mol Biol 2023; 435:167629. [PMID: 35595170 DOI: 10.1016/j.jmb.2022.167629] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023]
Abstract
Action potential-induced neurotransmitter release in presynaptic boutons involves coordinated actions of a large list of proteins that are associated directly or indirectly with membrane structures including synaptic vesicles and plasma membranes. These proteins are often highly abundant in different synaptic bouton sub-compartments, and they rarely act alone. Instead, these proteins interact with each other forming intricate and distinct molecular complexes. Many of these complexes form condensed clusters on membrane surfaces. This review summarizes findings in recent years showing that many of presynaptic protein complex assemblies are formed via phase separation. These protein condensates extensively interact with lipid membranes via distinct modes, forming various mesoscale structures by different mode of organizations between membraneless condensates and membranous organelles. We discuss that such mesoscale interactions could have deep implications on mobilization, exocytosis, and retrieval of synaptic vesicles.
Collapse
Affiliation(s)
- Xiandeng Wu
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hua Qiu
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518036, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
23
|
Gao V, Briano JA, Komer LE, Burré J. Functional and Pathological Effects of α-Synuclein on Synaptic SNARE Complexes. J Mol Biol 2023; 435:167714. [PMID: 35787839 PMCID: PMC10472340 DOI: 10.1016/j.jmb.2022.167714] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
α-Synuclein is an abundant protein at the neuronal synapse that has been implicated in Parkinson's disease for over 25 years and characterizes the hallmark pathology of a group of neurodegenerative diseases now known as the synucleinopathies. Physiologically, α-synuclein exists in an equilibrium between a synaptic vesicle membrane-bound α-helical multimer and a cytosolic largely unstructured monomer. Through its membrane-bound state, α-synuclein functions in neurotransmitter release by modulating several steps in the synaptic vesicle cycle, including synaptic vesicle clustering and docking, SNARE complex assembly, and homeostasis of synaptic vesicle pools. These functions have been ascribed to α-synuclein's interactions with the synaptic vesicle SNARE protein VAMP2/synaptobrevin-2, the synaptic vesicle-attached synapsins, and the synaptic vesicle membrane itself. How α-synuclein affects these processes, and whether disease is due to loss-of-function or gain-of-toxic-function of α-synuclein remains unclear. In this review, we provide an in-depth summary of the existing literature, discuss possible reasons for the discrepancies in the field, and propose a working model that reconciles the findings in the literature.
Collapse
Affiliation(s)
- Virginia Gao
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Department of Neurology, New York Presbyterian/Weill Cornell Medicine, New York, NY, USA.
| | - Juan A Briano
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lauren E Komer
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA. https://www.twitter.com/lauren_komer
| | - Jacqueline Burré
- Appel Alzheimer's Disease Research Institute & Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
24
|
Yan P, Liu H, Zhou T, Sun P, Wang Y, Wang X, Zhang L, Wang T, Dong J, Zhu J, Lv L, Li W, Qi S, Liang Y, Kong E. Crosstalk of Synapsin1 palmitoylation and phosphorylation controls the dynamicity of synaptic vesicles in neurons. Cell Death Dis 2022; 13:786. [PMID: 36097267 PMCID: PMC9468182 DOI: 10.1038/s41419-022-05235-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 01/21/2023]
Abstract
The dynamics of synaptic vesicles (SVs) within presynaptic domains are tightly controlled by synapsin1 phosphorylation; however, the mechanism underlying the anchoring of synapsin1 with F-actin or SVs is not yet fully understood. Here, we found that Syn1 is modified with protein palmitoylation, and examining the roles of Syn1 palmitoylation in neurons led us to uncover that Syn1 palmitoylation is negatively regulated by its phosphorylation; together, they manipulate the clustering and redistribution of SVs. Using the combined approaches of electron microscopy and genetics, we revealed that Syn1 palmitoylation is vital for its binding with F-actin but not SVs. Inhibition of Syn1 palmitoylation causes defects in SVs clustering and a reduced number of total SVs in vivo. We propose a model in which SVs redistribution is triggered by upregulated Syn1 phosphorylation and downregulated Syn1 palmitoylation, and they reversibly promote SVs clustering. The crosstalk of Syn1 palmitoylation and phosphorylation thereby bidirectionally manipulates SVs dynamics in neurons.
Collapse
Affiliation(s)
- Peipei Yan
- grid.412990.70000 0004 1808 322XThe Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China ,grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Huicong Liu
- grid.412990.70000 0004 1808 322XThe Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China ,grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Tao Zhou
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Pu Sun
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Yilin Wang
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Xibin Wang
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Lin Zhang
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Tian Wang
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Jing Dong
- grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| | - Jiangli Zhu
- grid.13291.380000 0001 0807 1581Department of Urology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, 610041 Chengdu, China
| | - Luxian Lv
- grid.412990.70000 0004 1808 322XThe Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- grid.412990.70000 0004 1808 322XThe Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shiqian Qi
- grid.13291.380000 0001 0807 1581Department of Urology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, 610041 Chengdu, China
| | - Yinming Liang
- grid.412990.70000 0004 1808 322XLaboratory of Genetic Regulators in the Immune System, Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Eryan Kong
- grid.412990.70000 0004 1808 322XThe Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China ,grid.412990.70000 0004 1808 322XInstitute of Psychiatry and Neuroscience, Xinxiang key laboratory of protein palmitoylation and major human diseases, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
25
|
Insulin-like growth factor 1 regulates excitatory synaptic transmission in pyramidal neurons from adult prefrontal cortex. Neuropharmacology 2022; 217:109204. [PMID: 35931212 DOI: 10.1016/j.neuropharm.2022.109204] [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: 04/28/2022] [Revised: 07/07/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022]
Abstract
Insulin-like growth factor 1 (IGF1) influences synaptic function in addition to its role in brain development and aging. Although the expression levels of IGF1 and IGF1 receptor (IGF1R) peak during development and decline with age, the adult brain has abundant IGF1 or IGF1R expression. Studies reveal that IGF1 regulates the synaptic transmission in neurons from young animals. However, the action of IGF1 on neurons in the adult brain is still unclear. Here, we used prefrontal cortical (PFC) slices from adult mice (∼8 weeks old) to characterize the role of IGF1 on excitatory synaptic transmission in pyramidal neurons and the underlying molecular mechanisms. We first validated IGF1R expression in pyramidal neurons using translating ribosomal affinity purification assay. Then, using whole-cell patch-clamp recording, we found that IGF1 attenuated the amplitude of evoked excitatory postsynaptic current (EPSC) without affecting the frequency and amplitude of miniature EPSC. Furthermore, this decrease in excitatory neurotransmission was blocked by pharmacological inhibition of IGF1R or conditionally knockdown of IGF1R in PFC pyramidal neurons. In addition, we determined that IGF1-induced decrease of EPSC amplitude was due to postsynaptic effect (internalization of a-amino-3-hydroxy-5-methyl-4- isoxazolepropionic acid receptors [AMPAR]) rather than presynaptic glutamate release. Finally, we found that inhibition of metabotropic glutamate receptor subtype-1 (mGluR1) abolished IGF1-induced attenuation of evoked EPSC amplitude and decrease of AMPAR expression at synaptic membrane, suggesting mGluR1-mediated endocytosis of AMPAR was involved. Taken together, these data provide the first evidence that IGF1 regulates excitatory synaptic transmission in adult PFC via the interaction between IGF1R-dependent signaling pathway and mGluR1-mediated AMPAR endocytosis.
Collapse
|
26
|
McNaughton R, Huo Y, Li G, Ioschpe ADV, Yan L, Man HY, Zhang X. Regulatory Effects of Gradient Microtopographies on Synapse Formation and Neurite Growth in Hippocampal Neurons. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2022; 32:075005. [PMID: 35814808 PMCID: PMC9262107 DOI: 10.1088/1361-6439/ac73d7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Among approaches aiming toward functional nervous system restoration, those implementing microfabrication techniques allow the manufacture of platforms with distinct geometry where neurons can develop and be guided to form patterned connections in vitro. The interplay between neuronal development and the microenvironment, shaped by the physical limitations, remains largely unknown. Therefore, it is crucial to have an efficient way to quantify neuronal morphological changes induced by physical or contact guidance of the microenvironment. In this study, we first devise and assess a method to prepare anisotropic, gradient poly(dimethylsiloxane) micro-ridge/groove arrays featuring variable local pattern width. We then demonstrate the ability of this single substrate to simultaneously profile the morphologcial and synaptic connectivity changes of primary cultured hippocampal neurons reacting to variable physical conditons, throughout neurodevelopment, in vitro. The gradient microtopography enhanced adhesion within microgrooves, increasing soma density with decreasing pattern width. Decreasing pattern width also reduced dendritic arborization and increased preferential axon growth. Finally, decreasing pattern geometry inhibited presynaptic puncta architecture. Collectively, a method to examine structural development and connectivity in response to physical stimuli is established, and potentially provides insight into microfabricated geometries which promote neural regeneration and repair.
Collapse
Affiliation(s)
- Ryan McNaughton
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Yuda Huo
- Department of Biology, Boston University, Boston, MA, USA
| | - Guicai Li
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | | | - Lei Yan
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Xin Zhang
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| |
Collapse
|
27
|
Pramipexole treatment attenuates mechanical hypersensitivity in male rats experiencing chronic inflammatory pain. Neuropharmacology 2022; 208:108976. [PMID: 35085583 PMCID: PMC10171400 DOI: 10.1016/j.neuropharm.2022.108976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/14/2022] [Accepted: 01/22/2022] [Indexed: 11/23/2022]
Abstract
Opioids are commonly prescribed for pain despite growing evidence of their low efficacy in the treatment of chronic inflammatory pain and the high potential for misuse. There is a clear need to investigate non-opioid alternatives for the treatment of pain. In the present study, we tested the hypothesis that acute and repeated dopamine agonist treatment would attenuate mechanical hypersensitivity in male Long-Evans rats experiencing chronic inflammatory pain. We used two clinically available therapeutics, l-DOPA (precursor of dopamine biosynthesis) and pramipexole (dopamine D2/3 receptor agonist), to examine the functional role of dopamine signaling on mechanical hypersensitivity using an animal model of chronic inflammatory pain (complete Freund's adjuvant, CFA). We found that both acute and repeated pramipexole treatment attenuated hyperalgesia-like behavior in CFA-treated animals but exhibited no analgesic effects in control animals. In contrast, there was no effect of acute or repeated l-DOPA treatment on mechanical hypersensitivity in either CFA- or saline-treated animals. Notably, we discovered some extended effects of l-DOPA and pramipexole on decreasing pain-like behavior at three days and one week post-drug treatment. We also examined the effects of pramipexole treatment on glutamatergic and presynaptic signaling in pain- and reward-related brain regions including the nucleus accumbens (NAc), dorsal striatum (DS), ventral tegmental area (VTA), cingulate cortex (CC), central amygdala (CeA), and periaqueductal gray (PAG). We found that pramipexole treatment decreased AMPA receptor phosphorylation (pGluR1845) in the NAc and DS but increased pGluR1845 in the CC and CeA. A marker of presynaptic vesicle release, pSynapsin, was also increased in the DS, VTA, CC, CeA, and PAG following pramipexole treatment. Interestingly, pramipexole increased pSynapsin in the NAc of saline-treated animals, but not CFA-treated animals, suggesting blunted presynaptic vesicle release in the NAc of CFA-treated animals following pramipexole treatment. To examine the functional implications of impaired presynaptic signaling in the NAc of CFA animals, we used ex vivo electrophysiology to examine the effects of pramipexole treatment on the intrinsic excitability of NAc neurons in CFA- and saline-treated animals. We found that pramipexole treatment reduced NAc intrinsic excitability in saline-treated animals but produced no change in NAc intrinsic excitability in CFA-treated animals. These findings indicate alterations in dopamine D2/3 receptor signaling in the NAc of animals with a history of chronic pain in association with the anti-hyperalgesic effects of pramipexole treatment.
Collapse
|
28
|
Moschetta M, Ravasenga T, De Fusco A, Maragliano L, Aprile D, Orlando M, Sacchetti S, Casagrande S, Lignani G, Fassio A, Baldelli P, Benfenati F. Ca 2+ binding to synapsin I regulates resting Ca 2+ and recovery from synaptic depression in nerve terminals. Cell Mol Life Sci 2022; 79:600. [PMID: 36409372 PMCID: PMC9678998 DOI: 10.1007/s00018-022-04631-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/23/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022]
Abstract
Synapsin I (SynI) is a synaptic vesicle (SV)-associated phosphoprotein that modulates neurotransmission by controlling SV trafficking. The SynI C-domain contains a highly conserved ATP binding site mediating SynI oligomerization and SV clustering and an adjacent main Ca2+ binding site, whose physiological role is unexplored. Molecular dynamics simulations revealed that the E373K point mutation irreversibly deletes Ca2+ binding to SynI, still allowing ATP binding, but inducing a destabilization of the SynI oligomerization interface. Here, we analyzed the effects of this mutation on neurotransmitter release and short-term plasticity in excitatory and inhibitory synapses from primary hippocampal neurons. Patch-clamp recordings showed an increase in the frequency of miniature excitatory postsynaptic currents (EPSCs) that was totally occluded by exogenous Ca2+ chelators and associated with a constitutive increase in resting terminal Ca2+ concentrations. Evoked EPSC amplitude was also reduced, due to a decreased readily releasable pool (RRP) size. Moreover, in both excitatory and inhibitory synapses, we observed a marked impaired recovery from synaptic depression, associated with impaired RRP refilling and depletion of the recycling pool of SVs. Our study identifies SynI as a novel Ca2+ buffer in excitatory terminals. Blocking Ca2+ binding to SynI results in higher constitutive Ca2+ levels that increase the probability of spontaneous release and disperse SVs. This causes a decreased size of the RRP and an impaired recovery from depression due to the failure of SV reclustering after sustained high-frequency stimulation. The results indicate a physiological role of Ca2+ binding to SynI in the regulation of SV clustering and trafficking in nerve terminals.
Collapse
Affiliation(s)
- Matteo Moschetta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Tiziana Ravasenga
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Antonio De Fusco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Davide Aprile
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy ,Present Address: High-Definition Disease Modelling Lab, Campus IFOM-IEO, Milan, Italy
| | - Marta Orlando
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Present Address: Charitè Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany
| | - Silvio Sacchetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Silvia Casagrande
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Gabriele Lignani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,Present Address: Queens Square Institute of Neurology, University College London, London, UK
| | - Anna Fassio
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano Di Tecnologia, Largo Rosanna Benzi 10, 16132 Genoa, Italy ,IRCCS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
29
|
Imbrosci B, Schmitz D, Orlando M. Automated Detection and Localization of Synaptic Vesicles in Electron Microscopy Images. eNeuro 2022; 9:ENEURO.0400-20.2021. [PMID: 34983830 PMCID: PMC8805189 DOI: 10.1523/eneuro.0400-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/04/2021] [Accepted: 11/23/2021] [Indexed: 11/21/2022] Open
Abstract
Information transfer and integration in the brain occurs at chemical synapses and is mediated by the fusion of synaptic vesicles filled with neurotransmitter. Synaptic vesicle dynamic spatial organization regulates synaptic transmission as well as synaptic plasticity. Because of their small size, synaptic vesicles require electron microscopy (EM) for their imaging, and their analysis is conducted manually. The manual annotation and segmentation of the hundreds to thousands of synaptic vesicles, is highly time consuming and limits the throughput of data collection. To overcome this limitation, we built an algorithm, mainly relying on convolutional neural networks (CNNs), capable of automatically detecting and localizing synaptic vesicles in electron micrographs. The algorithm was trained on murine synapses but we show that it works well on synapses from different species, ranging from zebrafish to human, and from different preparations. As output, we provide the vesicle count and coordinates, the nearest neighbor distance (nnd) and the estimate of the vesicles area. We also provide a graphical user interface (GUI) to guide users through image analysis, result visualization, and manual proof-reading. The application of our algorithm is especially recommended for images produced by transmission EM. Since this type of imaging is used routinely to investigate presynaptic terminals, our solution will likely be of interest for numerous research groups.
Collapse
Affiliation(s)
- Barbara Imbrosci
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience (BCCN) Berlin, Berlin 10115, Germany
- Einstein Center for Neurosciences (ECN) Berlin, Berlin 10117, Germany
- Max-Delbrück-Centrum (MDC) for Molecular Medicine, Berlin 13125, Germany
| | - Marta Orlando
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Berlin 10117, Germany
| |
Collapse
|
30
|
Presynaptic AMPA Receptors in Health and Disease. Cells 2021; 10:cells10092260. [PMID: 34571906 PMCID: PMC8470629 DOI: 10.3390/cells10092260] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
AMPA receptors (AMPARs) are ionotropic glutamate receptors that play a major role in excitatory neurotransmission. AMPARs are located at both presynaptic and postsynaptic plasma membranes. A huge number of studies investigated the role of postsynaptic AMPARs in the normal and abnormal functioning of the mammalian central nervous system (CNS). These studies highlighted that changes in the functional properties or abundance of postsynaptic AMPARs are major mechanisms underlying synaptic plasticity phenomena, providing molecular explanations for the processes of learning and memory. Conversely, the role of AMPARs at presynaptic terminals is as yet poorly clarified. Accruing evidence demonstrates that presynaptic AMPARs can modulate the release of various neurotransmitters. Recent studies also suggest that presynaptic AMPARs may possess double ionotropic-metabotropic features and that they are involved in the local regulation of actin dynamics in both dendritic and axonal compartments. In addition, evidence suggests a key role of presynaptic AMPARs in axonal pathology, in regulation of pain transmission and in the physiology of the auditory system. Thus, it appears that presynaptic AMPARs play an important modulatory role in nerve terminal activity, making them attractive as novel pharmacological targets for a variety of pathological conditions.
Collapse
|
31
|
Longhena F, Faustini G, Brembati V, Pizzi M, Benfenati F, Bellucci A. An updated reappraisal of synapsins: structure, function and role in neurological and psychiatric disorders. Neurosci Biobehav Rev 2021; 130:33-60. [PMID: 34407457 DOI: 10.1016/j.neubiorev.2021.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/29/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023]
Abstract
Synapsins (Syns) are phosphoproteins strongly involved in neuronal development and neurotransmitter release. Three distinct genes SYN1, SYN2 and SYN3, with elevated evolutionary conservation, have been described to encode for Synapsin I, Synapsin II and Synapsin III, respectively. Syns display a series of common features, but also exhibit distinctive localization, expression pattern, post-translational modifications (PTM). These characteristics enable their interaction with other synaptic proteins, membranes and cytoskeletal components, which is essential for the proper execution of their multiple functions in neuronal cells. These include the control of synapse formation and growth, neuron maturation and renewal, as well as synaptic vesicle mobilization, docking, fusion, recycling. Perturbations in the balanced expression of Syns, alterations of their PTM, mutations and polymorphisms of their encoding genes induce severe dysregulations in brain networks functions leading to the onset of psychiatric or neurological disorders. This review presents what we have learned since the discovery of Syn I in 1977, providing the state of the art on Syns structure, function, physiology and involvement in central nervous system disorders.
Collapse
Affiliation(s)
- Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Viviana Brembati
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Fabio Benfenati
- Italian Institute of Technology, Via Morego 30, Genova, Italy; IRCSS Policlinico San Martino Hospital, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy; Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| |
Collapse
|
32
|
Hoffmann C, Sansevrino R, Morabito G, Logan C, Vabulas RM, Ulusoy A, Ganzella M, Milovanovic D. Synapsin Condensates Recruit alpha-Synuclein. J Mol Biol 2021; 433:166961. [PMID: 33774037 DOI: 10.1016/j.jmb.2021.166961] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/07/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022]
Abstract
Neurotransmission relies on the tight spatial and temporal regulation of the synaptic vesicle (SV) cycle. Nerve terminals contain hundreds of SVs that form tight clusters. These clusters represent a distinct liquid phase in which one component of the phase are SVs and the other synapsin 1, a highly abundant synaptic protein. Another major family of disordered proteins at the presynapse includes synucleins, most notably α-synuclein. The precise physiological role of α-synuclein in synaptic physiology remains elusive, albeit its role has been implicated in nearly all steps of the SV cycle. To determine the effect of α-synuclein on the synapsin phase, we employ the reconstitution approach using natively purified SVs from rat brains and the heterologous cell system to generate synapsin condensates. We demonstrate that synapsin condensates recruit α-synuclein, and while enriched into these synapsin condensates, α-synuclein still maintains its high mobility. The presence of SVs enhances the rate of synapsin/α-synuclein condensation, suggesting that SVs act as catalyzers for the formation of synapsin condensates. Notably, at physiological salt and protein concentrations, α-synuclein alone is not able to cluster isolated SVs. Excess of α-synuclein disrupts the kinetics of synapsin/SV condensate formation, indicating that the molar ratio between synapsin and α-synuclein is important in assembling the functional condensates of SVs. Understanding the molecular mechanism of α-synuclein interactions at the nerve terminals is crucial for clarifying the pathogenesis of synucleinopathies, where α-synuclein, synaptic proteins and lipid organelles all accumulate as insoluble intracellular inclusions.
Collapse
Affiliation(s)
- Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Roberto Sansevrino
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Giuseppe Morabito
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Chinyere Logan
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - R Martin Vabulas
- Charité - Universitätsmedizin Berlin, Institute of Biochemistry, 10117 Berlin, Germany
| | - Ayse Ulusoy
- Laboratory of Neuroprotective Mechanisms, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Marcelo Ganzella
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany.
| |
Collapse
|
33
|
Pannuzzo M. Beta-amyloid pore linked to controlled calcium influx into the cell: A new paradigm for Alzheimer's Disease. Alzheimers Dement 2021; 18:191-196. [PMID: 34051062 PMCID: PMC9290490 DOI: 10.1002/alz.12373] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/20/2021] [Accepted: 04/16/2021] [Indexed: 11/28/2022]
Abstract
Despite tremendous worldwide efforts, clinical trials assessing Alzheimer's disease (AD)‐related therapeutics have been relentlessly unsuccessful. Hence, there is an urgent need to challenge old hypotheses with novel paradigms. An emerging concept is that the amyloid‐beta (Aβ) peptide, which was until recently deemed a major player in the cause of AD, may instead modulate synaptic plasticity and protect against excitotoxicity. The link between Aβ‐mediated synaptic plasticity and Aβ trafficking is central for understanding AD pathogenesis and remains a perplexing relationship. The crossover between Aβ pathological and physiological roles is subtle and remains controversial. Based on existing literature, as a signaling molecule, Aβ is proposed to modulate its own turnover and synaptic plasticity through what is currently believed to be the cause of AD: the transient formation of pore‐like oligomers. A change of perspective regarding how Aβ pores exert a protective function will unavoidably revolutionize the entire field of anti‐amyloid drug development.
Collapse
Affiliation(s)
- Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
34
|
Runwal G, Edwards RH. The Membrane Interactions of Synuclein: Physiology and Pathology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:465-485. [PMID: 33497259 DOI: 10.1146/annurev-pathol-031920-092547] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Specific proteins accumulate in neurodegenerative disease, and human genetics has indicated a causative role for many. In most cases, however, the mechanisms remain poorly understood. Degeneration is thought to involve a gain of abnormal function, although we do not know the normal function of many proteins implicated. The protein α-synuclein accumulates in the Lewy pathology of Parkinson's disease and related disorders, and mutations in α-synuclein cause degeneration, but we have not known its normal function or how it triggers disease. α-Synuclein localizes to presynaptic boutons and interacts with membranes in vitro. Overexpression slows synaptic vesicle exocytosis, and recent data suggest a normal role for the endogenous synucleins in dilation of the exocytic fusion pore. Disrupted membranes also appear surprisingly prominent in Lewy pathology. Synuclein thus interacts with membranes under both physiological and pathological conditions, suggesting that the normal function of synuclein may illuminate its role in degeneration.
Collapse
Affiliation(s)
- Gautam Runwal
- Departments of Neurology and Physiology, Graduate Programs in Cell Biology, Biomedical Sciences and Neuroscience, School of Medicine, University of California, San Francisco, California 94143, USA;
| | - Robert H Edwards
- Departments of Neurology and Physiology, Graduate Programs in Cell Biology, Biomedical Sciences and Neuroscience, School of Medicine, University of California, San Francisco, California 94143, USA;
| |
Collapse
|
35
|
Zhang M, Augustine GJ. Synapsins and the Synaptic Vesicle Reserve Pool: Floats or Anchors? Cells 2021; 10:cells10030658. [PMID: 33809712 PMCID: PMC8002314 DOI: 10.3390/cells10030658] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 11/24/2022] Open
Abstract
In presynaptic terminals, synaptic vesicles (SVs) are found in a discrete cluster that includes a reserve pool that is mobilized during synaptic activity. Synapsins serve as a key protein for maintaining SVs within this reserve pool, but the mechanism that allows synapsins to do this is unclear. This mechanism is likely to involve synapsins either cross-linking SVs, thereby anchoring SVs to each other, or creating a liquid phase that allows SVs to float within a synapsin droplet. Here, we summarize what is known about the role of synapsins in clustering of SVs and evaluate experimental evidence supporting these two models.
Collapse
|
36
|
Snca-GFP Knock-In Mice Reflect Patterns of Endogenous Expression and Pathological Seeding. eNeuro 2020; 7:ENEURO.0007-20.2020. [PMID: 32788297 PMCID: PMC7470929 DOI: 10.1523/eneuro.0007-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/06/2020] [Accepted: 06/13/2020] [Indexed: 12/21/2022] Open
Abstract
α-Synuclein (aSyn) participates in synaptic vesicle trafficking and synaptic transmission but its misfolding is also strongly implicated in Parkinson’s disease (PD) and other neurodegenerative synucleinopathies in which misfolded aSyn accumulates in different regions of the central and peripheral nervous systems. Although increased aSyn expression levels or altered aggregation propensities likely underlie familial PD with SNCA amplification or mutations, the majority of synucleinopathies arise sporadically, indicating that disease can develop under normal levels of wild-type (wt) aSyn. We report here the development and characterization of a mouse line expressing an aSyn-green fluorescence protein (GFP) fusion protein under the control of native Snca regulatory elements. Regional and subcellular localization of the aSyn-GFP fusion protein in brains and peripheral tissues of knock-in (KI) mice are indistinguishable from that of wt littermates. Importantly, similar to wt aSyn, aSyn-GFP disperses from synaptic vesicles on membrane depolarization, indicating that the tag does not alter normal aSyn dynamics at synapses. In addition, intracerebral injection of aSyn pre-formed fibrils into KI mice induced the formation of aSyn-GFP inclusions with a distribution pattern similar to that observed in wt mice, albeit with attenuated kinetics because of the GFP-tag. We anticipate that this new mouse model will facilitate in vitro and in vivo studies requiring in situ detection of endogenous aSyn, thereby providing new insights into aSyn function in health and disease.
Collapse
|
37
|
The Synaptic Vesicle Cycle Revisited: New Insights into the Modes and Mechanisms. J Neurosci 2020; 39:8209-8216. [PMID: 31619489 DOI: 10.1523/jneurosci.1158-19.2019] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 02/01/2023] Open
Abstract
Neurotransmission is sustained by endocytosis and refilling of synaptic vesicles (SVs) locally within the presynapse. Until recently, a consensus formed that after exocytosis, SVs are recovered by either fusion pore closure (kiss-and-run) or clathrin-mediated endocytosis directly from the plasma membrane. However, recent data have revealed that SV formation is more complex than previously envisaged. For example, two additional recycling pathways have been discovered, ultrafast endocytosis and activity-dependent bulk endocytosis, in which SVs are regenerated from the internalized membrane and synaptic endosomes. Furthermore, these diverse modes of endocytosis appear to influence both the molecular composition and subsequent physiological role of individual SVs. In addition, previously unknown complexity in SV refilling and reclustering has been revealed. This review presents a modern view of the SV life cycle and discusses how neuronal subtype, physiological temperature, and individual activity patterns can recruit different endocytic modes to generate new SVs and sculpt subsequent presynaptic performance.
Collapse
|
38
|
Abstract
Emerging evidence indicates that liquid-liquid phase separation, the formation of a condensed molecular assembly within another diluted aqueous solution, is a means for cells to organize highly condensed biological assemblies (also known as biological condensates or membraneless compartments) with very broad functions and regulatory properties in different subcellular regions. Molecular machineries dictating synaptic transmissions in both presynaptic boutons and postsynaptic densities of neuronal synapses may be such biological condensates. Here we review recent developments showing how phase separation can build dense synaptic molecular clusters, highlight unique features of such condensed clusters in the context of synaptic development and signaling, discuss how aberrant phase-separation-mediated synaptic assembly formation may contribute to dysfunctional signaling in psychiatric disorders, and present some challenges and opportunities of phase separation in synaptic biology.
Collapse
|
39
|
Pechstein A, Tomilin N, Fredrich K, Vorontsova O, Sopova E, Evergren E, Haucke V, Brodin L, Shupliakov O. Vesicle Clustering in a Living Synapse Depends on a Synapsin Region that Mediates Phase Separation. Cell Rep 2020; 30:2594-2602.e3. [DOI: 10.1016/j.celrep.2020.01.092] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/15/2019] [Accepted: 01/24/2020] [Indexed: 12/28/2022] Open
|
40
|
Merino P, Diaz A, Torre ER, Yepes M. Urokinase-type plasminogen activator (uPA) regulates the expression and function of growth-associated protein 43 (GAP-43) in the synapse. J Biol Chem 2019; 295:619-630. [PMID: 31819012 DOI: 10.1074/jbc.ra119.010644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/19/2019] [Indexed: 11/06/2022] Open
Abstract
Growth-associated protein 43 (GAP-43) plays a central role in the formation of presynaptic terminals, synaptic plasticity, and axonal growth and regeneration. During development, GAP-43 is found in axonal extensions of most neurons. In contrast, in the mature brain, its expression is restricted to a few presynaptic terminals and scattered axonal growth cones. Urokinase-type plasminogen activator (uPA) is a serine proteinase that, upon binding to its receptor (uPAR), catalyzes the conversion of plasminogen into plasmin and activates signaling pathways that promote cell migration, proliferation, and survival. In the developing brain, uPA induces neuritogenesis and neuronal migration. In contrast, the expression and function of uPA in the mature brain are poorly understood. However, recent evidence reveals that different forms of injury induce release of uPA and expression of uPAR in neurons and that uPA/uPAR binding triggers axonal growth and synapse formation. Here we show that binding of uPA to uPAR induces not only the mobilization of GAP-43 from the axonal shaft to the presynaptic terminal but also its activation in the axonal bouton by PKC-induced calcium-dependent phosphorylation at Ser-41 (pGAP-43). We found that this effect requires open presynaptic N-methyl-d-aspartate receptors but not plasmin generation. Furthermore, our work reveals that, following its activation by uPA/uPAR binding, pGAP-43 colocalizes with presynaptic vesicles and triggers their mobilization to the synaptic release site. Together, these data reveal a novel role of uPA as an activator of the synaptic vesicle cycle in cerebral cortical neurons via its ability to induce presynaptic recruitment and activation of GAP-43.
Collapse
Affiliation(s)
- Paola Merino
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208
| | - Ariel Diaz
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208
| | - Enrique R Torre
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208
| | - Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, Georgia 30329-4208; Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322-0001; Department of Neurology, Veterans Affairs Medical Center, Atlanta, Georgia 30033-4004.
| |
Collapse
|
41
|
Sulzer D, Edwards RH. The physiological role of α-synuclein and its relationship to Parkinson's Disease. J Neurochem 2019; 150:475-486. [PMID: 31269263 PMCID: PMC6707892 DOI: 10.1111/jnc.14810] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/03/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022]
Abstract
The protein α-synuclein has a central role in the pathogenesis of Parkinson's disease (PD). In this review, we discuss recent results concerning its primary function, which appears to be on cell membranes. The pre-synaptic location of synuclein has suggested a role in neurotransmitter release and it apparently associates with synaptic vesicles because of their high curvature. Indeed, synuclein over-expression inhibits synaptic vesicle exocytosis. However, loss of synuclein has not yet been shown to have a major effect on synaptic transmission. Consistent with work showing that synuclein can promote as well as sense membrane curvature, recent analysis of synuclein triple knockout mice now shows that synuclein accelerates dilation of the exocytic fusion pore. This form of regulation affects primarily the release of slowly discharged lumenal cargo such as neural peptides, but presumably also contributes to maintenance of the release site. This article is part of the Special Issue "Synuclein".
Collapse
Affiliation(s)
- David Sulzer
- Departments of Psychiatry, Neurology and Pharmacology, Columbia University Medical Center, New York State Psychiatric Institute
| | - Robert H Edwards
- Departments of Neurology and Physiology, Graduate Programs in Cell Biology, Biomedical Sciences and Neuroscience, UCSF School of Medicine
| |
Collapse
|
42
|
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: 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: 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
|
43
|
Vaden JH, Banumurthy G, Gusarevich ES, Overstreet-Wadiche L, Wadiche JI. The readily-releasable pool dynamically regulates multivesicular release. eLife 2019; 8:47434. [PMID: 31364987 PMCID: PMC6716946 DOI: 10.7554/elife.47434] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/30/2019] [Indexed: 01/04/2023] Open
Abstract
The number of neurotransmitter-filled vesicles released into the synaptic cleft with each action potential dictates the reliability of synaptic transmission. Variability of this fundamental property provides diversity of synaptic function across brain regions, but the source of this variability is unclear. The prevailing view is that release of a single (univesicular release, UVR) or multiple vesicles (multivesicular release, MVR) reflects variability in vesicle release probability, a notion that is well-supported by the calcium-dependence of release mode. However, using mouse brain slices, we now demonstrate that the number of vesicles released is regulated by the size of the readily-releasable pool, upstream of vesicle release probability. Our results point to a model wherein protein kinase A and its vesicle-associated target, synapsin, dynamically control release site occupancy to dictate the number of vesicles released without altering release probability. Together these findings define molecular mechanisms that control MVR and functional diversity of synaptic signaling. Our nervous system allows us to rapidly sense and respond to the world around us via cells called neurons that relay electrical signals around the brain and body. When an electrical impulse travelling along one neuron reaches a junction – called a synapse – with a neighboring neuron, it stimulates small containers known as vesicles from the first cell to release their contents into the synapse. These contents then travel across to the neighboring cell and may generate a new electrical impulse. The number of vesicles at a synapse that are ready to be released varies from one to ten. The more vesicles the neuron releases, the more likely the second cell will produce an electrical signal of its own. However, not all electrical signals reaching a synapse stimulate vesicles to be released and some signals only release a single vesicle. What determines how many vesicles are released by a single electrical signal? Some vesicles have a higher likelihood of being released than others, but this “eagerness” does not always predict how many vesicles an individual synapse will actually discharge. Now, Vaden et al. have used brain tissue from mice to test an alternative possibility: the simple idea that the number of vesicles available at the synapse affects how many vesicles are released without altering their eagerness for release. Vaden et al. found that activating an enzyme called protein kinase A increased the number of vesicles released from synapses without changing how likely individual vesicles were to be released. Inhibiting protein kinase A also did not change individual vesicle’s eagerness to be released, but did decrease the number of vesicles that were discharged. Further experiments found that protein kinase A modifies a molecule on the surface of vesicles, known as synapsin, which controls the number of vesicles that are available for release. These findings show that the number of vesicles released at a synapse is controlled by two independently regulated parameters: the number of vesicles that are available, as well as how eager individual vesicles are to be released. The ability of neurons to communicate with each other is disrupted in autism spectrum disorders, Alzheimer’s disease and many other diseases. Learning how neurons communicate in healthy brains will help us understand what happens in the neurons of individuals with these conditions.
Collapse
Affiliation(s)
- Jada H Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | | | - Eugeny S Gusarevich
- Department of Fundamental and Applied Physics, Northern (Arctic) Federal University named after M.V. Lomonosov, Arkhangelsk, Russian Federation
| | | | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| |
Collapse
|
44
|
Marte A, Russo I, Rebosio C, Valente P, Belluzzi E, Pischedda F, Montani C, Lavarello C, Petretto A, Fedele E, Baldelli P, Benfenati F, Piccoli G, Greggio E, Onofri F. Leucine‐rich repeat kinase 2 phosphorylation on synapsin I regulates glutamate release at pre‐synaptic sites. J Neurochem 2019; 150:264-281. [PMID: 31148170 DOI: 10.1111/jnc.14778] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 05/20/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain scaffolding protein with kinase and GTPase activities involved in synaptic vesicle (SV) dynamics. While its role in Parkinson's disease has been largely investigated, little is known about LRRK2 physiological role and until now few proteins have been described as substrates. We have previously demonstrated that LRRK2 through its WD40 domain interacts with synapsin I, an important SV-associated phosphoprotein involved in neuronal development and in the regulation of neurotransmitter release. To test whether synapsin I is substrate for LRRK2 and characterize the properties of its phosphorylation, we used in vitro kinase and binding assays as well as cellular model and site-direct mutagenesis. Using synaptosomes in superfusion, patch-clamp recordings in autaptic WT and synapsin I KO cortical neurons and SypHy assay on primary cortical culture from wild-type and BAC human LRRK2 G2019S mice we characterized the role of LRRK2 kinase activity on glutamate release and SV trafficking. Here we reported that synapsin I is phosphorylated by LRRK2 and demonstrated that the interaction between LRRK2 WD40 domain and synapsin I is crucial for this phosphorylation. Moreover, we showed that LRRK2 phosphorylation of synapsin I at threonine 337 and 339 significantly reduces synapsin I-SV/actin interactions. Using complementary experimental approaches, we demonstrated that LRRK2 controls glutamate release and SV dynamics in a kinase activity and synapsin I-dependent manner. Our findings show that synapsin I is a LRRK2 substrate and describe a novel mechanisms of regulation of glutamate release by LRRK2 kinase activity.
Collapse
Affiliation(s)
- Antonella Marte
- Department of Experimental Medicine University of Genova Genova Italy
| | | | | | - Pierluigi Valente
- Department of Experimental Medicine University of Genova Genova Italy
- IRCCS Ospedale Policlinico San Martino Genova Italy
| | - Elisa Belluzzi
- Rheumatology Unit, Department of Medicine‐DIMED University Hospital of Padova Padova Italy
| | - Francesca Pischedda
- Center for Integrative Biology (CIBIO) University of Trento Trento Italy
- Dulbecco Telethon Institute Trento Italy
| | - Caterina Montani
- Center for Integrative Biology (CIBIO) University of Trento Trento Italy
- Dulbecco Telethon Institute Trento Italy
| | - Chiara Lavarello
- Laboratory of Mass Spectrometry ‐ Core Facilities Istituto Giannina Gaslini Genova Italy
| | - Andrea Petretto
- Laboratory of Mass Spectrometry ‐ Core Facilities Istituto Giannina Gaslini Genova Italy
| | - Ernesto Fedele
- Department of Pharmacy University of Genova Genova Italy
- IRCCS Ospedale Policlinico San Martino Genova Italy
| | - Pietro Baldelli
- Department of Experimental Medicine University of Genova Genova Italy
- IRCCS Ospedale Policlinico San Martino Genova Italy
| | - Fabio Benfenati
- IRCCS Ospedale Policlinico San Martino Genova Italy
- Center for Synaptic Neuroscience and Technology Istituto Italiano di Tecnologia Genova Italy
| | - Giovanni Piccoli
- Center for Integrative Biology (CIBIO) University of Trento Trento Italy
- Dulbecco Telethon Institute Trento Italy
| | - Elisa Greggio
- Department of Biology University of Padova Padova Italy
| | - Franco Onofri
- Department of Experimental Medicine University of Genova Genova Italy
- IRCCS Ospedale Policlinico San Martino Genova Italy
| |
Collapse
|
45
|
SIV-Mediated Synaptic Dysfunction Is Associated with an Increase in Synapsin Site 1 Phosphorylation and Impaired PP2A Activity. J Neurosci 2019; 39:7006-7018. [PMID: 31270156 DOI: 10.1523/jneurosci.0178-19.2019] [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: 01/19/2019] [Revised: 05/31/2019] [Accepted: 06/22/2019] [Indexed: 11/21/2022] Open
Abstract
Although the reduction of viral loads in people with HIV undergoing combination antiretroviral therapy has mitigated AIDS-related symptoms, the prevalence of neurological impairments has remained unchanged. HIV-associated CNS dysfunction includes impairments in memory, attention, memory processing, and retrieval. Here, we show a significant site-specific increase in the phosphorylation of Syn I serine 9, site 1, in the frontal cortex lysates and synaptosome preparations of male rhesus macaques infected with simian immunodeficiency virus (SIV) but not in uninfected or SIV-infected antiretroviral therapy animals. Furthermore, we found that a lower protein phosphatase 2A (PP2A) activity, a phosphatase responsible for Syn I (S9) dephosphorylation, is primarily associated with the higher S9 phosphorylation in the frontal cortex of SIV-infected macaques. Comparison of brain sections confirmed higher Syn I (S9) in the frontal cortex and greater coexpression of Syn I and PP2A A subunit, which was observed as perinuclear aggregates in the somata of the frontal cortex of SIV-infected macaques. Synaptosomes from SIV-infected animals were physiologically tested using a synaptic vesicle endocytosis assay and FM4-64 dye showing a significantly higher baseline depolarization levels in synaptosomes of SIV+-infected than uninfected control or antiretroviral therapy animals. A PP2A-activating FDA-approved drug, FTY720, decreased the higher synaptosome depolarization in SIV-infected animals. Our results suggest that an impaired distribution and lower activity of serine/threonine phosphatases in the context of HIV infection may cause an indirect effect on the phosphorylation levels of essential proteins involving in synaptic transmission, supporting the occurrence of specific impairments in the synaptic activity during SIV infection.SIGNIFICANCE STATEMENT Even with antiretroviral therapy, neurocognitive deficits, including impairments in attention, memory processing, and retrieval, are still major concerns in people living with HIV. Here, we used the rhesus macaque simian immunodeficiency virus model with and without antiretroviral therapy to study the dynamics of phosphorylation of key amino acid residues of synapsin I, which critically impacts synaptic vesicle function. We found a significant increase in synapsin I phosphorylation at serine 9, which was driven by dysfunction of serine/threonine protein phosphatase 2A in the nerve terminals. Our results suggest that an impaired distribution and lower activity of serine/threonine phosphatases in the context of HIV infection may cause an indirect effect on the phosphorylation levels of essential proteins involved in synaptic transmission.
Collapse
|
46
|
Mikheeva IB, Malkov AE, Pavlik LL, Arkhipov VI, Levin SG. Effect of TGF-beta1 on long-term synaptic plasticity and distribution of AMPA receptors in the CA1 field of the hippocampus. Neurosci Lett 2019; 704:95-99. [PMID: 30953737 DOI: 10.1016/j.neulet.2019.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/06/2019] [Accepted: 04/02/2019] [Indexed: 11/29/2022]
Abstract
Using the methods of electrophysiology and immunohistochemistry, the effect of the transforming factor beta-1(TGF-β1), an anti-inflammatory cytokine, on the long-term post-tetanic potentiation (LTP) in CA1 field hippocampal slices and the distribution of the GluR1 subunit of the AMPA receptor has been studied. It was shown that TGF-β1 at a concentration of 10 ng/ml did not significantly affect the initial stage of LTP and substantially changed the distribution of synaptic AMPA receptors in response to tetanic stimulation. Twenty five minutes after the tetanization, the main pool of AMPA receptors (90%) was due to the postsynaptic density (PSD). By contrast, LTP in the presence of TGF-β1 was accompanied by less pronounced changes in the distribution of AMPA receptors. Their localization in both pre- and postsynaptic regions remained nearly the same as that in the control. It may be suggested that the normal distribution of AMPA receptors in spinous synapses promotes the stabilization of potentiated synapses, thereby retaining LTP for longer terms.
Collapse
Affiliation(s)
- I B Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Pushchino State Institute of Natural Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - A E Malkov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - L L Pavlik
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - V I Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Pushchino State Institute of Natural Sciences, Pushchino, Moscow Region, 142290, Russia
| | - S G Levin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Pushchino State Institute of Natural Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
47
|
Fu T, Wang J, Ding Y, Zhang Y, Han S, Li J. Modulation of cPKCγ on Synapsin-Ia/b-Specific Phosphorylation Sites in the Developing Visual Cortex of Mice. Invest Ophthalmol Vis Sci 2019; 60:2676-2684. [PMID: 31242289 DOI: 10.1167/iovs.19-26675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To explore the role of synapsin-Ia/b in visual cortical plasticity, the dynamic changes in total protein expression (T-) and conventional protein kinase C (cPKC)γ-modulated phosphorylation (P-) levels of synapsin-Ia/b were observed in the developing visual cortex of mice. Methods The Western blot analysis was used to determine the levels of T- and P-synapsin-Ia/b at site of Ser9, 549, and 603; the cPKCγ gene wild-type (cPKCγ+/+) and knockout (cPKCγ-/-) mice were applied to explore the modulation of cPKCγ on synapsin-Ia/b phosphorylation status in visual cortex of mice at postnatal 7 to 60 days (P7-P60, n = 6 per group). Results The results showed that T-synapsin-Ia/b protein levels significantly increased at P14 to P35 and peaked at P42 to 60 (P < 0.001) in visual cortex when compared with that of P7 cPKCγ+/+ mice, and cPKCγ-/- did not affect this pattern of T-synapsin-Ia/b protein expressions. For synapsin-Ia/b phosphorylation status, the levels of P-Ser9 and 603 synapsin-Ia/b significantly elevated at P21 to P28 (P < 0.05 or 0.001), and then went down and maintained at lower levels at P35 to P60 (P < 0.05 or 0.001) compared with P7 cPKCγ+/+ mice. In addition, the cPKCγ gene knockout could significantly (P < 0.001) inhibit both the increase and decrease of P-Ser9 and 603 synapsin-Ia/b levels when compared with cPKCγ+/+ mice at P7 to P60. However, there were no significant changes of P-Ser549 synapsin-Ia/b in the developing visual cortex of both cPKCγ+/+ and cPKCγ-/- mice at P7 to P60. Conclusions These results suggested that both protein expression levels and cPKCγ-modulated phosphorylation status at Ser9 and 603 of synapsin-Ia/b may play important role in developing visual cortex of mice.
Collapse
Affiliation(s)
- Tao Fu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Jing Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Yichao Ding
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Yunxia Zhang
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Song Han
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
48
|
Silm K, Yang J, Marcott PF, Asensio CS, Eriksen J, Guthrie DA, Newman AH, Ford CP, Edwards RH. Synaptic Vesicle Recycling Pathway Determines Neurotransmitter Content and Release Properties. Neuron 2019; 102:786-800.e5. [PMID: 31003725 PMCID: PMC6541489 DOI: 10.1016/j.neuron.2019.03.031] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 01/28/2019] [Accepted: 03/19/2019] [Indexed: 01/03/2023]
Abstract
In contrast to temporal coding by synaptically acting neurotransmitters such as glutamate, neuromodulators such as monoamines signal changes in firing rate. The two modes of signaling have been thought to reflect differences in release by different cells. We now find that midbrain dopamine neurons release glutamate and dopamine with different properties that reflect storage in different synaptic vesicles. The vesicles differ in release probability, coupling to presynaptic Ca2+ channels and frequency dependence. Although previous work has attributed variation in these properties to differences in location or cytoskeletal association of synaptic vesicles, the release of different transmitters shows that intrinsic differences in vesicle identity drive different modes of release. Indeed, dopamine but not glutamate vesicles depend on the adaptor protein AP-3, revealing an unrecognized linkage between the pathway of synaptic vesicle recycling and the properties of exocytosis. Storage of the two transmitters in different vesicles enables the transmission of distinct signals.
Collapse
Affiliation(s)
- Kätlin Silm
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Jing Yang
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Pamela F Marcott
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Cedric S Asensio
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Jacob Eriksen
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Daryl A Guthrie
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes of Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Amy H Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes of Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Christopher P Ford
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert H Edwards
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA.
| |
Collapse
|
49
|
He WY, Zhang B, Zhao WC, He J, Zhang L, Xiong QM, Wang J, Wang HB. Contributions of mTOR Activation-Mediated Upregulation of Synapsin II and Neurite Outgrowth to Hyperalgesia in STZ-Induced Diabetic Rats. ACS Chem Neurosci 2019; 10:2385-2396. [PMID: 30785256 DOI: 10.1021/acschemneuro.8b00680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Painful diabetic neuropathy (PDN) is among the common complications in diabetes mellitus (DM), with its underlying mechanisms largely unknown. Synapsin II is primarily expressed in the spinal dorsal horn, and its upregulation mediates a superfluous release of glutamate and a deficiency of GABAergic interneuron synaptic transmission, which is directly implicated in the facilitation of pain signals in the hyperalgesic nociceptive response. Recently, synapsin II has been revealed to be associated with the modulation of neurite outgrowth, whereas the process of this neuronal structural neuroplasticity following neuronal hyperexcitability still remains unclear. In this study, we found that under conditions of elevated glucose, TNF-α induced the activation of mTOR, mediating the upregulation of synapsin II and neurite outgrowth in dorsal horn neurons. In vivo, we demonstrated that mTOR and synapsin II were upregulated and coexpressed in the spinal dorsal horn neurons in rats with streptozotocin (STZ)-induced diabetes. Furthermore, the intrathecal administration of the mTOR inhibitor rapamycin or synapsin II shRNA significantly diminished the expression of synapsin II, effectively mitigating hyperalgesia in PDN rats. We are the first to discover that in STZ-induced diabetic rats the activation of mTOR mediates the upregulation of synapsin II and neurite outgrowth, both contributing to hyperalgesia. These findings may benefit the clinical therapy of PDN by provision of a novel target.
Collapse
Affiliation(s)
- Wan-you He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Bin Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Wei-cheng Zhao
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Jian He
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Lei Zhang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Qing-ming Xiong
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Jing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| | - Han-bing Wang
- Department of Anesthesiology, The First People’s Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, China
| |
Collapse
|
50
|
Farizatto KLG, Almeida MF, Long RT, Bahr BA. Early Synaptic Alterations and Selective Adhesion Signaling in Hippocampal Dendritic Zones Following Organophosphate Exposure. Sci Rep 2019; 9:6532. [PMID: 31024077 PMCID: PMC6484076 DOI: 10.1038/s41598-019-42934-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/11/2019] [Indexed: 11/09/2022] Open
Abstract
Organophosphates account for many of the world's deadliest poisons. They inhibit acetylcholinesterase causing cholinergic crises that lead to seizures and death, while survivors commonly experience long-term neurological problems. Here, we treated brain explants with the organophosphate compound paraoxon and uncovered a unique mechanism of neurotoxicity. Paraoxon-exposed hippocampal slice cultures exhibited progressive declines in synaptophysin, synapsin II, and PSD-95, whereas reduction in GluR1 was slower and NeuN and Nissl staining showed no indications of neuronal damage. The distinctive synaptotoxicity was observed in dendritic zones of CA1 and dentate gyrus. Interestingly, declines in synapsin II dendritic labeling correlated with increased staining for β1 integrin, a component of adhesion receptors that regulate synapse maintenance and plasticity. The paraoxon-induced β1 integrin response was targeted to synapses, and the two-fold increase in β1 integrin was selective as other synaptic adhesion molecules were unchanged. Additionally, β1 integrin-cofilin signaling was triggered by the exposure and correlations were found between the extent of synaptic decline and the level of β1 integrin responses. These findings identified organophosphate-mediated early and lasting synaptotoxicity which can explain delayed neurological dysfunction later in life. They also suggest that the interplay between synaptotoxic events and compensatory adhesion responses influences neuronal fate in exposed individuals.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ronald T Long
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.
| |
Collapse
|