1
|
Pati E, Franceschi Biagioni A, Casani R, Lozano N, Kostarelos K, Cellot G, Ballerini L. Delivery of graphene oxide nanosheets modulates glutamate release and normalizes amygdala synaptic plasticity to improve anxiety-related behavior. NANOSCALE 2023; 15:18581-18591. [PMID: 37955642 DOI: 10.1039/d3nr04490d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Graphene oxide nanosheets (GO) were reported to alter neurobiological processes involving cell membrane dynamics. GO ability to reversibly downregulate specifically glutamatergic synapses underpins their potential in future neurotherapeutic developments. Aberrant glutamate plasticity contributes to stress-related psychopathology and drugs which target dysregulated glutamate represent promising treatments. We find that in a rat model of post-traumatic stress disorder (PTSD), a single injection of GO to the lateral amygdala following the stressful event induced PTSD-related behavior remission and reduced dendritic spine densities. We explored from a mechanistic perspective how GO could impair glutamate synaptic plasticity. By simultaneous patch clamp pair recordings of unitary synaptic currents, live-imaging of presynaptic vesicle release and confocal microscopy, we report that GO nanosheets altered the probability of release enhancing the extinction of synaptic plasticity in the amygdala. These findings show that the modulation of presynaptic glutamate release might represent an unexplored target for (nano)pharmacological interventions of stress-related disorders.
Collapse
Affiliation(s)
- Elisa Pati
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| | | | - Raffaele Casani
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| | - Neus Lozano
- Nanomedicine Lab Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Kostas Kostarelos
- Nanomedicine Lab Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Nanomedicine Lab, and Faculty of Biology, Medicine & Health, The National Graphene Institute, University of Manchester, Manchester, M13 9PL, UK
| | - Giada Cellot
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| | - Laura Ballerini
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| |
Collapse
|
2
|
Lu CW, Lin TY, Hsieh PW, Chiu KM, Lee MY, Wang SJ. Cynarin, a caffeoylquinic acid derivative in artichoke, inhibits exocytotic glutamate release from rat cortical nerve terminals (synaptosomes). Neurochem Int 2023; 167:105537. [PMID: 37164158 DOI: 10.1016/j.neuint.2023.105537] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
The purpose of this study was to evaluate the effect of cynarin, a caffeoylquinic acid derivative in artichoke, on glutamate release elicited by 4-aminopyridine (4-AP) in rat cortical nerve terminals (synaptosomes). We observed that cynarin decreased 4-aminopyridine-elicited glutamate release, which was prevented by the removal of external free Ca2+ with ethylene glycol bis (β-aminoethyl ether)-N,N,N,N-tetraacetic acid (EGTA) or the blockade of P/Q-type calcium channels with ω-agatoxin IVA. Molecular docking also revealed that cynarin formed a hydrogen bond with the P/Q-type Ca2+ channel, indicating a mechanism of action involving Ca2+ influx inhibition. Additionally, the inhibitory effect of cynarin on glutamate release is associated with a change in the available synaptic vesicles, as cynarin decreased 4-AP-elicited FM1-43 release or hypertonic sucrose-evoked glutamate release from synaptosomes. Furthermore, the suppression of protein kinase A (PKA) prevented the effect of cynarin on 4-AP-elicited glutamate release. 4-AP-elicited PKA and synapsin I or synaptosomal-associated protein of 25 kDa (SNAP-25) phosphorylation at PKA-specific residues were also attenuated by cynarin. Our data indicate that cynarin, through the suppression of P/Q-type Ca2+ channels, inhibits PKA activation and attenuates synapsin I and SNAP-25 phosphorylation at PKA-specific residues, thus decreasing synaptic vesicle availability and contributing to glutamate release inhibition in cerebral cortex terminals.
Collapse
Affiliation(s)
- Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Pei-Wen Hsieh
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, And Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33303, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei, 22060, Taiwan
| | - Su-Jane Wang
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan.
| |
Collapse
|
3
|
Hsu SK, Lu CW, Chiu KM, Lee MY, Lin TY, Wang SJ. Mangiferin depresses vesicular glutamate release in synaptosomes from the rat cerebral cortex by decreasing synapsin I phosphorylation. Eur J Pharmacol 2023; 950:175772. [PMID: 37146708 DOI: 10.1016/j.ejphar.2023.175772] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
Mangiferin is a glucosyl xanthone that has been shown to be a neuroprotective agent against brain disorders involving excess glutamate. However, the effect of mangiferin on the function of the glutamatergic system has not been investigated. In this study, we used synaptosomes from the rat cerebral cortex to investigate the effect of mangiferin on glutamate release and identify the possible underlying mechanism. We observed that mangiferin produced a concentration-dependent reduction in the release of glutamate elicited by 4-aminopyridine with an IC50 value of 25 μM. Inhibition of glutamate release was blocked by removing extracellular calcium and by treatment with the vacuolar-type H+-ATPase inhibitor bafilomycin A1, which prevents the uptake and storage of glutamate in vesicles. Moreover, we showed that mangiferin decreased the 4-aminopyridine-elicited FM1-43 release and synaptotagmin 1 luminal domain antibody (syt1-L ab) uptake from synaptosomes, which correlated with decreased synaptic vesicle exocytosis. Transmission electron microscopy in synaptosomes also showed that mangiferin attenuated the 4-aminopyridine-elicited decrease in the number of synaptic vesicles. In addition, antagonism of Ca2+/calmodulin-dependent kinase II (CaMKII) and protein kinase A (PKA) counteracted mangiferin's effect on glutamate release. Mangiferin also decreased the phosphorylation of CaMKII, PKA, and synapsin I elicited by 4-aminopyridine treatment. Our data suggest that mangiferin reduces PKA and CaMKII activation and synapsin I phosphorylation, which could decrease synaptic vesicle availability and lead to a subsequent reduction in vesicular glutamate release from synaptosomes.
Collapse
Affiliation(s)
- Szu-Kai Hsu
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Department of Neurosurgery, Cathay General Hospital, Taipei, 106438, Taiwan
| | - Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, 33303, Taiwan.
| |
Collapse
|
4
|
Biswal S, Wasnik V. Accuracy in readout of glutamate concentrations by neuronal cells. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:30. [PMID: 37076657 DOI: 10.1140/epje/s10189-023-00287-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/28/2023] [Indexed: 05/03/2023]
Abstract
Glutamate and glycine are important neurotransmitters in the brain. An action potential propagating in the terminal of a presynaptic neuron causes the release of glutamate and glycine in the synapse by vesicles fusing with the cell membrane, which then activate various receptors on the cell membrane of the post-synaptic neuron. Entry of Ca[Formula: see text] through the activated NMDA receptors leads to a host of cellular processes of which long-term potentiation is of crucial importance because it is widely considered to be one of the major mechanisms behind learning and memory. By analysing the readout of glutamate concentration by the post-synaptic neurons during Ca[Formula: see text] signaling, we find that the average receptor density in hippocampal neurons has evolved to allow for accurate measurement of the glutamate concentration in the synaptic cleft.
Collapse
Affiliation(s)
- Swoyam Biswal
- Indian Institute of Technology Goa At Goa College of Engineering Campus, Farmagudi, Ponda-403401, Goa, India
| | - Vaibhav Wasnik
- Indian Institute of Technology Goa At Goa College of Engineering Campus, Farmagudi, Ponda-403401, Goa, India.
| |
Collapse
|
5
|
Krishnan S, Klingauf J. The readily retrievable pool of synaptic vesicles. Biol Chem 2023; 404:385-397. [PMID: 36867726 DOI: 10.1515/hsz-2022-0298] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023]
Abstract
In the CNS communication between neurons occurs at synapses by secretion of neurotransmitter via exocytosis of synaptic vesicles (SVs) at the active zone. Given the limited number of SVs in presynaptic boutons a fast and efficient recycling of exocytosed membrane and proteins by triggered compensatory endocytosis is required to maintain neurotransmission. Thus, pre-synapses feature a unique tight coupling of exo- and endocytosis in time and space resulting in the reformation of SVs with uniform morphology and well-defined molecular composition. This rapid response requires early stages of endocytosis at the peri-active zone to be well choreographed to ensure reformation of SVs with high fidelity. The pre-synapse can address this challenge by a specialized membrane microcompartment, where a pre-sorted and pre-assembled readily retrievable pool (RRetP) of endocytic membrane patches is formed, consisting of the vesicle cargo, presumably bound within a nucleated Clathrin and adaptor complex. This review considers evidence for the RRetP microcompartment to be the primary organizer of presynaptic triggered compensatory endocytosis.
Collapse
Affiliation(s)
- Sai Krishnan
- Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch Strasse 31, D-48149, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch Strasse 31, D-48149, Münster, Germany.,Center for Soft Nanoscience, Busso-Peus Strasse 10, D-48149, Münster, Germany
| |
Collapse
|
6
|
Biasetti L, Rey S, Fowler M, Ratnayaka A, Fennell K, Smith C, Marshall K, Hall C, Vargas-Caballero M, Serpell L, Staras K. Elevated amyloid beta disrupts the nanoscale organization and function of synaptic vesicle pools in hippocampal neurons. Cereb Cortex 2023; 33:1263-1276. [PMID: 35368053 PMCID: PMC9930632 DOI: 10.1093/cercor/bhac134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 11/14/2022] Open
Abstract
Alzheimer's disease is linked to increased levels of amyloid beta (Aβ) in the brain, but the mechanisms underlying neuronal dysfunction and neurodegeneration remain enigmatic. Here, we investigate whether organizational characteristics of functional presynaptic vesicle pools, key determinants of information transmission in the central nervous system, are targets for elevated Aβ. Using an optical readout method in cultured hippocampal neurons, we show that acute Aβ42 treatment significantly enlarges the fraction of functional vesicles at individual terminals. We observe the same effect in a chronically elevated Aβ transgenic model (APPSw,Ind) using an ultrastructure-function approach that provides detailed information on nanoscale vesicle pool positioning. Strikingly, elevated Aβ is correlated with excessive accumulation of recycled vesicles near putative endocytic sites, which is consistent with deficits in vesicle retrieval pathways. Using the glutamate reporter, iGluSnFR, we show that there are parallel functional consequences, where ongoing information signaling capacity is constrained. Treatment with levetiracetam, an antiepileptic that dampens synaptic hyperactivity, partially rescues these transmission defects. Our findings implicate organizational and dynamic features of functional vesicle pools as targets in Aβ-driven synaptic impairment, suggesting that interventions to relieve the overloading of vesicle retrieval pathways might have promising therapeutic value.
Collapse
Affiliation(s)
- Luca Biasetti
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| | - Stephanie Rey
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
- National Physical Laboratory, Middlesex, TW11 0LW, United Kingdom
| | - Milena Fowler
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| | - Arjuna Ratnayaka
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
- Faculty of Medicine, University of Southampton, SO17 1BJ, United Kingdom
| | - Kate Fennell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| | - Catherine Smith
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| | - Karen Marshall
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| | - Catherine Hall
- Sussex Neuroscience, School of Psychology, University of Sussex, Brighton, BN1 9QH, United Kingdom
| | - Mariana Vargas-Caballero
- School of Biological Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, United Kingdom
| | - Louise Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| | - Kevin Staras
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom
| |
Collapse
|
7
|
Abstract
Precise and efficient coupling of endocytosis to exocytosis is critical for neurotransmission. The activity-dependent facilitation of endocytosis has been well established for efficient membrane retrieval; however, whether neural activity clamps endocytosis to avoid excessive membrane retrieval remains debatable with the mechanisms largely unknown. The present work provides compelling evidence that synaptotagmin-1 (Syt1) functions as a primary bidirectional Ca2+ sensor to promote slow, small-sized clathrin-mediated endocytosis but inhibit the fast, large-sized bulk endocytosis during elevated neural activity, the disruption of which leads to inefficient vesicle recycling under mild stimulation but excessive membrane retrieval following sustained neurotransmission. Thus, Syt1 serves as a fine-tuning Ca2+ sensor to ensure both efficient and precise coupling of endocytosis to exocytosis in response to different neural activities. Exocytosis and endocytosis are tightly coupled. In addition to initiating exocytosis, Ca2+ plays critical roles in exocytosis–endocytosis coupling in neurons and nonneuronal cells. Both positive and negative roles of Ca2+ in endocytosis have been reported; however, Ca2+ inhibition in endocytosis remains debatable with unknown mechanisms. Here, we show that synaptotagmin-1 (Syt1), the primary Ca2+ sensor initiating exocytosis, plays bidirectional and opposite roles in exocytosis–endocytosis coupling by promoting slow, small-sized clathrin-mediated endocytosis but inhibiting fast, large-sized bulk endocytosis. Ca2+-binding ability is required for Syt1 to regulate both types of endocytic pathways, the disruption of which leads to inefficient vesicle recycling under mild stimulation and excessive membrane retrieval following intense stimulation. Ca2+-dependent membrane tubulation may explain the opposite endocytic roles of Syt1 and provides a general membrane-remodeling working model for endocytosis determination. Thus, Syt1 is a primary bidirectional Ca2+ sensor facilitating clathrin-mediated endocytosis but clamping bulk endocytosis, probably by manipulating membrane curvature to ensure both efficient and precise coupling of endocytosis to exocytosis.
Collapse
|
8
|
Petrov AM, Zakirjanova GF, Kovyazina IV, Tsentsevitsky AN, Bukharaeva EA. Adrenergic receptors control frequency-dependent switching of the exocytosis mode between "full-collapse" and "kiss-and-run" in murine motor nerve terminal. Life Sci 2022; 296:120433. [PMID: 35219696 DOI: 10.1016/j.lfs.2022.120433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022]
Abstract
AIMS Neurotransmitter release from the synaptic vesicles can occur through two modes of exocytosis: "full-collapse" or "kiss-and-run". Here we investigated how increasing the nerve activity and pharmacological stimulation of adrenoceptors can influence the mode of exocytosis in the motor nerve terminal. METHODS Recording of endplate potentials with intracellular microelectrodes was used to estimate acetylcholine release. A fluorescent dye FM1-43 and its quenching with sulforhodamine 101 were utilized to visualize synaptic vesicle recycling. KEY FINDINGS An increase in the frequency of stimulation led to a decrease in the rate of FM1-43 unloading despite the higher number of quanta released. High frequency activity promoted neurotransmitter release via the kiss-and-run mechanism. This was confirmed by experiments utilizing (I) FM1-43 dye quencher, that is able to pass into the synaptic vesicle via fusion pore, and (II) loading of FM1-43 by compensatory endocytosis. Noradrenaline and specific α2-adrenoreceptors agonist, dexmedetomidine, controlled the mode of synaptic vesicle recycling at high frequency activity. Their applications favored neurotransmitter release via full-collapse exocytosis rather than the kiss-and-run pathway. SIGNIFICANCE At the diaphragm neuromuscular junctions, neuronal commands are translated into contractions necessary for respiration. During stress, an increase in discharge rate of the phrenic nerve shifts the exocytosis from the full-collapse to the kiss-and-run mode. The stress-related molecule, noradrenaline, restricts neurotransmitter release in response to a high frequency activity, and prevents the shift in the mode of exocytosis through α2-adrenoceptor activation. This may be a component of the mechanism that limits overstimulation of the respiratory system during stress.
Collapse
Affiliation(s)
- Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia; Kazan State Medical University, Kazan, Russia.
| | - Guzalia F Zakirjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia
| | - Irina V Kovyazina
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia; Kazan State Medical University, Kazan, Russia
| | - Andrei N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia
| | - Ellya A Bukharaeva
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, Russia
| |
Collapse
|
9
|
An SJ, Stagi M, Gould TJ, Wu Y, Mlodzianoski M, Rivera-Molina F, Toomre D, Strittmatter SM, De Camilli P, Bewersdorf J, Zenisek D. Multimodal imaging of synaptic vesicles with a single probe. CELL REPORTS METHODS 2022; 2:100199. [PMID: 35497490 PMCID: PMC9046237 DOI: 10.1016/j.crmeth.2022.100199] [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] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/11/2022] [Accepted: 03/28/2022] [Indexed: 05/17/2023]
Abstract
A complete understanding of synaptic-vesicle recycling requires the use of multiple microscopy methods to obtain complementary information. However, many currently available probes are limited to a specific microscopy modality, which necessitates the use of multiple probes and labeling paradigms. Given the complexity of vesicle populations and recycling pathways, having new single-vesicle probes that could be used for multiple microscopy techniques would complement existing sets of tools for studying vesicle function. Here, we present a probe based on the membrane-binding C2 domain of cytosolic phospholipase A2 (cPLA2) that fulfills this need. By conjugating the C2 domain with different detectable tags, we demonstrate that a single, modular probe can allow synaptic vesicles to be imaged at multiple levels of spatial and temporal resolution. Moreover, as a general endocytic marker, the C2 domain may also be used to study membrane recycling in many cell types.
Collapse
Affiliation(s)
- Seong J. An
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Massimiliano Stagi
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool 69 3BX, UK
| | - Travis J. Gould
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Physics and Astronomy, Bates College, Lewiston, ME 04240, USA
| | - Yumei Wu
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Michael Mlodzianoski
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Felix Rivera-Molina
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Derek Toomre
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen M. Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pietro De Camilli
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - David Zenisek
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
10
|
CDH2 mutation affecting N-cadherin function causes attention-deficit hyperactivity disorder in humans and mice. Nat Commun 2021; 12:6187. [PMID: 34702855 PMCID: PMC8548587 DOI: 10.1038/s41467-021-26426-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/29/2021] [Indexed: 11/20/2022] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a common childhood-onset psychiatric disorder characterized by inattention, impulsivity and hyperactivity. ADHD exhibits substantial heritability, with rare monogenic variants contributing to its pathogenesis. Here we demonstrate familial ADHD caused by a missense mutation in CDH2, which encodes the adhesion protein N-cadherin, known to play a significant role in synaptogenesis; the mutation affects maturation of the protein. In line with the human phenotype, CRISPR/Cas9-mutated knock-in mice harboring the human mutation in the mouse ortholog recapitulated core behavioral features of hyperactivity. Symptoms were modified by methylphenidate, the most commonly prescribed therapeutic for ADHD. The mutated mice exhibited impaired presynaptic vesicle clustering, attenuated evoked transmitter release and decreased spontaneous release. Specific downstream molecular pathways were affected in both the ventral midbrain and prefrontal cortex, with reduced tyrosine hydroxylase expression and dopamine levels. We thus delineate roles for CDH2-related pathways in the pathophysiology of ADHD. Molecular mechanisms of attention-deficit hyperactivity disorder (ADHD) are not fully understood. Here the authors demonstrate a mutation in CDH2, encoding N-cadherin, that is associated with ADHD, and in a mouse model, delineate molecular electrophysiological characteristics associated with this mutation.
Collapse
|
11
|
Preformed Ω-profile closure and kiss-and-run mediate endocytosis and diverse endocytic modes in neuroendocrine chromaffin cells. Neuron 2021; 109:3119-3134.e5. [PMID: 34411513 DOI: 10.1016/j.neuron.2021.07.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/02/2021] [Accepted: 07/23/2021] [Indexed: 01/29/2023]
Abstract
Transformation of flat membrane into round vesicles is generally thought to underlie endocytosis and produce speed-, amount-, and vesicle-size-specific endocytic modes. Visualizing depolarization-induced exocytic and endocytic membrane transformation in live neuroendocrine chromaffin cells, we found that flat membrane is transformed into Λ-shaped, Ω-shaped, and O-shaped vesicles via invagination, Λ-base constriction, and Ω-pore constriction, respectively. Surprisingly, endocytic vesicle formation is predominantly from not flat-membrane-to-round-vesicle transformation but calcium-triggered and dynamin-mediated closure of (1) Ω profiles formed before depolarization and (2) fusion pores (called kiss-and-run). Varying calcium influxes control the speed, number, and vesicle size of these pore closures, resulting in speed-specific slow (more than ∼6 s), fast (less than ∼6 s), or ultrafast (<0.6 s) endocytosis, amount-specific compensatory endocytosis (endocytosis = exocytosis) or overshoot endocytosis (endocytosis > exocytosis), and size-specific bulk endocytosis. These findings reveal major membrane transformation mechanisms underlying endocytosis, diverse endocytic modes, and exocytosis-endocytosis coupling, calling for correction of the half-a-century concept that the flat-to-round transformation predominantly mediates endocytosis after physiological stimulation.
Collapse
|
12
|
Jeon J, Yoon SH, Oh MA, Cho W, Kim JY, Shin CI, Kim EJ, Chung TD. Neuroligin-1-Modified Electrodes for Specific Coupling with a Presynaptic Neuronal Membrane. ACS APPLIED MATERIALS & INTERFACES 2021; 13:21944-21953. [PMID: 33909393 DOI: 10.1021/acsami.1c01298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Coordination of synapses onto electrodes with high specificity and maintaining a stable and long-lasting interface have importance in the field of neural interfaces. One potential approach is to present ligands on the surface of electrodes that would be bound through a protein-protein interaction to specific areas of neuronal cells. Here, we functionalize electrode surfaces with genetically engineered neuroligin-1 protein and demonstrate the formation of a nascent presynaptic bouton upon binding to neurexin-1 β on the presynaptic membrane of neurons. The resulting synaptically connected electrode shows an assembly of presynaptic proteins and comparable exocytosis kinetics to that of native synapses. Importantly, a neuroligin-1-induced synapse-electrode interface exhibits type specificity and structural robustness. We envision that the use of synaptic adhesion proteins in modified neural electrodes may lead to new approaches in the interfacing of neural circuity and electronics.
Collapse
Affiliation(s)
- Joohee Jeon
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Sun-Heui Yoon
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Min-Ah Oh
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Wonkyung Cho
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Yong Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Chang Il Shin
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Joong Kim
- Advanced Institute of Convergence Technology, Suwon-Si 16229, Gyeonggi-do, Republic of Korea
| | - Taek Dong Chung
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Advanced Institute of Convergence Technology, Suwon-Si 16229, Gyeonggi-do, Republic of Korea
| |
Collapse
|
13
|
Rey S, Marra V, Smith C, Staras K. Nanoscale Remodeling of Functional Synaptic Vesicle Pools in Hebbian Plasticity. Cell Rep 2021; 30:2006-2017.e3. [PMID: 32049027 PMCID: PMC7016504 DOI: 10.1016/j.celrep.2020.01.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/21/2019] [Accepted: 01/14/2020] [Indexed: 12/23/2022] Open
Abstract
Vesicle pool properties are known determinants of synaptic efficacy, but their potential role as modifiable substrates in forms of Hebbian plasticity is still unclear. Here, we investigate this using a nanoscale readout of functionally recycled vesicles in natively wired hippocampal CA3→CA1 circuits undergoing long-term potentiation (LTP). We show that the total recycled vesicle pool is larger after plasticity induction, with the smallest terminals exhibiting the greatest relative expansion. Changes in the spatial organization of vesicles accompany potentiation including a specific increase in the number of recycled vesicles at the active zone, consistent with an ultrastructural remodeling component of synaptic strengthening. The cAMP-PKA pathway activator, forskolin, selectively mimics some features of LTP-driven changes, suggesting that distinct and independent modules of regulation accompany plasticity expression. Our findings provide evidence for a presynaptic locus of LTP encoded in the number and arrangement of functionally recycled vesicles, with relevance for models of long-term plasticity storage.
Collapse
Affiliation(s)
- Stephanie Rey
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Vincenzo Marra
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester L1 7RH, United Kingdom
| | - Catherine Smith
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Kevin Staras
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom.
| |
Collapse
|
14
|
Brill SE, Maraslioglu A, Kurz C, Kramer F, Fuhr MF, Singh A, Friauf E. Glycinergic Transmission in the Presence and Absence of Functional GlyT2: Lessons From the Auditory Brainstem. Front Synaptic Neurosci 2021; 12:560008. [PMID: 33633558 PMCID: PMC7900164 DOI: 10.3389/fnsyn.2020.560008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Synaptic transmission is controlled by re-uptake systems that reduce transmitter concentrations in the synaptic cleft and recycle the transmitter into presynaptic terminals. The re-uptake systems are thought to ensure cytosolic concentrations in the terminals that are sufficient for reloading empty synaptic vesicles (SVs). Genetic deletion of glycine transporter 2 (GlyT2) results in severely disrupted inhibitory neurotransmission and ultimately to death. Here we investigated the role of GlyT2 at inhibitory glycinergic synapses in the mammalian auditory brainstem. These synapses are tuned for resilience, reliability, and precision, even during sustained high-frequency stimulation when endocytosis and refilling of SVs probably contribute substantially to efficient replenishment of the readily releasable pool (RRP). Such robust synapses are formed between MNTB and LSO neurons (medial nucleus of the trapezoid body, lateral superior olive). By means of patch-clamp recordings, we assessed the synaptic performance in controls, in GlyT2 knockout mice (KOs), and upon acute pharmacological GlyT2 blockade. Via computational modeling, we calculated the reoccupation rate of empty release sites and RRP replenishment kinetics during 60-s challenge and 60-s recovery periods. Control MNTB-LSO inputs maintained high fidelity neurotransmission at 50 Hz for 60 s and recovered very efficiently from synaptic depression. During 'marathon-experiments' (30,600 stimuli in 20 min), RRP replenishment accumulated to 1,260-fold. In contrast, KO inputs featured severe impairments. For example, the input number was reduced to ~1 (vs. ~4 in controls), implying massive functional degeneration of the MNTB-LSO microcircuit and a role of GlyT2 during synapse maturation. Surprisingly, neurotransmission did not collapse completely in KOs as inputs still replenished their small RRP 80-fold upon 50 Hz | 60 s challenge. However, they totally failed to do so for extended periods. Upon acute pharmacological GlyT2 inactivation, synaptic performance remained robust, in stark contrast to KOs. RRP replenishment was 865-fold in marathon-experiments, only ~1/3 lower than in controls. Collectively, our empirical and modeling results demonstrate that GlyT2 re-uptake activity is not the dominant factor in the SV recycling pathway that imparts indefatigability to MNTB-LSO synapses. We postulate that additional glycine sources, possibly the antiporter Asc-1, contribute to RRP replenishment at these high-fidelity brainstem synapses.
Collapse
Affiliation(s)
- Sina E Brill
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Ayse Maraslioglu
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Catharina Kurz
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Florian Kramer
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Martin F Fuhr
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Abhyudai Singh
- Electrical & Computer Engineering, University of Delaware, Newark, DE, United States
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
15
|
Ritzau-Jost A, Tsintsadze T, Krueger M, Ader J, Bechmann I, Eilers J, Barbour B, Smith SM, Hallermann S. Large, Stable Spikes Exhibit Differential Broadening in Excitatory and Inhibitory Neocortical Boutons. Cell Rep 2021; 34:108612. [PMID: 33440142 PMCID: PMC7809622 DOI: 10.1016/j.celrep.2020.108612] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 10/19/2020] [Accepted: 12/06/2020] [Indexed: 01/09/2023] Open
Abstract
Presynaptic action potential spikes control neurotransmitter release and thus interneuronal communication. However, the properties and the dynamics of presynaptic spikes in the neocortex remain enigmatic because boutons in the neocortex are small and direct patch-clamp recordings have not been performed. Here, we report direct recordings from boutons of neocortical pyramidal neurons and interneurons. Our data reveal rapid and large presynaptic action potentials in layer 5 neurons and fast-spiking interneurons reliably propagating into axon collaterals. For in-depth analyses, we establish boutons of mature cultured neurons as models for excitatory neocortical boutons, demonstrating that the presynaptic spike amplitude is unaffected by potassium channels, homeostatic long-term plasticity, and high-frequency firing. In contrast to the stable amplitude, presynaptic spikes profoundly broaden during high-frequency firing in layer 5 pyramidal neurons, but not in fast-spiking interneurons. Thus, our data demonstrate large presynaptic spikes and fundamental differences between excitatory and inhibitory boutons in the neocortex.
Collapse
Affiliation(s)
- Andreas Ritzau-Jost
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Timur Tsintsadze
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, OR 97239, USA; Section of Pulmonary and Critical Care Medicine, VA Portland Health Care System, Portland, OR 97239, USA
| | - Martin Krueger
- Institute of Anatomy, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Jonas Ader
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Jens Eilers
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Boris Barbour
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Université Paris, 75005 Paris, France
| | - Stephen M Smith
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, OR 97239, USA; Section of Pulmonary and Critical Care Medicine, VA Portland Health Care System, Portland, OR 97239, USA.
| | - Stefan Hallermann
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany.
| |
Collapse
|
16
|
Chenouard N, Xuan F, Tsien RW. Synaptic vesicle traffic is supported by transient actin filaments and regulated by PKA and NO. Nat Commun 2020; 11:5318. [PMID: 33087709 PMCID: PMC7578807 DOI: 10.1038/s41467-020-19120-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/25/2020] [Indexed: 11/10/2022] Open
Abstract
Synaptic vesicles (SVs) can be pooled across multiple synapses, prompting questions about their dynamic allocation for neurotransmission and plasticity. We find that the axonal traffic of recycling vesicles is not supported by ubiquitous microtubule-based motility but relies on actin instead. Vesicles freed from synaptic clusters undergo ~1 µm bouts of active transport, initiated by nearby elongation of actin filaments. Long distance translocation arises when successive bouts of active transport were linked by periods of free diffusion. The availability of SVs for active transport can be promptly increased by protein kinase A, a key player in neuromodulation. Vesicle motion is in turn impeded by shutting off axonal actin polymerization, mediated by nitric oxide-cyclic GMP signaling leading to inhibition of RhoA. These findings provide a potential framework for coordinating post-and pre-synaptic strength, using retrograde regulation of axonal actin dynamics to mobilize and recruit presynaptic SV resources.
Collapse
Affiliation(s)
- Nicolas Chenouard
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, 10016, USA.,Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Feng Xuan
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, 10016, USA.,Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, 60208, USA
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, 10016, USA. .,Center for Neural Science, New York University, New York, NY, 10003, USA.
| |
Collapse
|
17
|
Sakimoto Y, Mizuno J, Kida H, Kamiya Y, Ono Y, Mitsushima D. Learning Promotes Subfield-Specific Synaptic Diversity in Hippocampal CA1 Neurons. Cereb Cortex 2020; 29:2183-2195. [PMID: 30796817 PMCID: PMC6459007 DOI: 10.1093/cercor/bhz022] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
The hippocampus is functionally heterogeneous between the dorsal and ventral subfields with left–right asymmetry. To determine the possible location of contextual memory, we performed an inhibitory avoidance task to analyze synaptic plasticity using slice patch-clamp technique. The training bilaterally increased the AMPA/NMDA ratio at dorsal CA3–CA1 synapses, whereas the training did not affect the ratio at ventral CA3–CA1 synapses regardless of the hemisphere. Moreover, sequential recording of miniature excitatory postsynaptic currents and miniature inhibitory postsynaptic currents from the same CA1 neuron clearly showed learning-induced synaptic plasticity. In dorsal CA1 neurons, the training dramatically strengthened both excitatory and inhibitory postsynaptic responses in both hemispheres, whereas the training did not promote the plasticity in either hemisphere in ventral CA1 neurons. Nonstationary fluctuation analysis further revealed that the training bilaterally increased the number of AMPA or GABAA receptor channels at dorsal CA1 synapses, but not at ventral CA1 synapses, suggesting functional heterogeneity of learning-induced receptor mobility. Finally, the performance clearly impaired by the bilateral microinjection of plasticity blockers in dorsal, but not ventral CA1 subfields, suggesting a crucial role for contextual learning. The quantification of synaptic diversity in specified CA1 subfields may help us to diagnose and evaluate cognitive disorders at the information level.
Collapse
Affiliation(s)
- Y Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | | - H Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Y Kamiya
- Uonuma Institute of Community Medicine, Niigata University Medical Hospital, Niigata, Japan
| | - Y Ono
- Department of Electronics and Bioinformatics, Meiji University School of Science and Technology, Tokyo, Japan
| | - D Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.,Department of Physiology and Neuroscience, Kanagawa Dental University, Kanagawa, Japan.,The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
18
|
Sanderson TM, Georgiou J, Collingridge GL. Illuminating Relationships Between the Pre- and Post-synapse. Front Neural Circuits 2020; 14:9. [PMID: 32308573 PMCID: PMC7146027 DOI: 10.3389/fncir.2020.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Excitatory synapses in the mammalian cortex are highly diverse, both in terms of their structure and function. However, relationships between synaptic features indicate they are highly coordinated entities. Imaging techniques, that enable physiology at the resolution of individual synapses to be investigated, have allowed the presynaptic activity level of the synapse to be related to postsynaptic function. This approach has revealed that neuronal activity induces the pre- and post-synapse to be functionally correlated and that subsets of synapses are more susceptible to certain forms of synaptic plasticity. As presynaptic function is often examined in isolation from postsynaptic properties, the effect it has on the post-synapse is not fully understood. However, since postsynaptic receptors at excitatory synapses respond to release of glutamate, it follows that they may be differentially regulated depending on the frequency of its release. Therefore, examining postsynaptic properties in the context of presynaptic function may be a useful way to approach a broad range of questions on synaptic physiology. In this review, we focus on how optophysiology tools have been utilized to study relationships between the pre- and the post-synapse. Multiple imaging techniques have revealed correlations in synaptic properties from the submicron to the dendritic level. Optical tools together with advanced imaging techniques are ideally suited to illuminate this area further, due to the spatial resolution and control they allow.
Collapse
Affiliation(s)
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Graham L Collingridge
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, University of Toronto, Toronto, ON, Canada.,Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
19
|
Sakimoto Y, Kida H, Mitsushima D. Temporal dynamics of learning-promoted synaptic diversity in CA1 pyramidal neurons. FASEB J 2019; 33:14382-14393. [PMID: 31689120 PMCID: PMC6894079 DOI: 10.1096/fj.201801893rrr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although contextual learning requires plasticity at both excitatory and inhibitory (E/I) synapses in cornu ammonis 1 (CA1) neurons, the temporal dynamics across the neuronal population are poorly understood. Using an inhibitory avoidance task, we analyzed the dynamic changes in learning-induced E/I synaptic plasticity. The training strengthened GABAA receptor–mediated synapses within 1 min, peaked at 10 min, and lasted for over 60 min. The intracellular loop (Ser408−409) of GABAA receptor β3 subunit was also phosphorylated within 1 min of training. As the results of strengthening of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor–mediated synapses, CA1 pyramidal neurons exhibited broad diversity of E/I synaptic currents within 5 min. Moreover, presynaptic glutamate release probability at basal dendrites also increased within 5 min. To further quantify the diversified E/I synaptic currents, we calculated self-entropy (bit) for individual neurons. The neurons showed individual levels of the parameter, which rapidly increased within 1 min of training and maintained for over 60 min. These results suggest that learning-induced synaptic plasticity is critical immediately following encoding rather than during the retrieval phase of the learning. Understanding the temporal dynamics along with the quantification of synaptic diversity would be necessary to identify a failure point for learning-promoted plasticity in cognitive disorders.—Sakimoto, Y., Kida, H., Mitsushima, D. Temporal dynamics of learning-promoted synaptic diversity in CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Yuya Sakimoto
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Hiroyuki Kida
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Dai Mitsushima
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan.,The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
20
|
Bilkey J, Nahirney PC, Delaney KR. Time and exposure to serotonin affect releasability of recycled vesicles at crayfish claw opener muscle synapses. Synapse 2019; 74:e22136. [PMID: 31574172 DOI: 10.1002/syn.22136] [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: 08/09/2019] [Revised: 09/16/2019] [Accepted: 09/21/2019] [Indexed: 11/10/2022]
Abstract
The crayfish claw opener neuromuscular junction is a biological model for studying presynaptic neuromodulation by serotonin (5HT) and synaptic vesicle recycling. It has been hypothesized that 5HT enhances release by recruiting a population of either previously nonrecycling or "reluctant" vesicles to increase the readily releasable pool. To determine if 5HT activates a distinct population of synaptic vesicles, recycling membranes were labeled with the membrane dye, FM1-43. Unloading (destaining) protocols could not resolve a population of vesicles that were only releasable in the presence of 5HT. Instead, we conclude synaptic vesicles change behavior in axon terminals independent of 5HT, becoming less likely to exocytose and unload dye over periods of >1 hr after recycling. We hypothesized this to be due to the slow conversion of a portion of recycled vesicles to a difficult to release state. The possibility that vesicles in these pools were spatially separated within the terminal was tested using photoconversion of FM1-43 and transmission electron microscopy. The location of FM1-43-labeled vesicles fixed 2 min following 3 min of 20-Hz stimulation did not reveal preferential localization of recycling vesicles specifically near release sites and the distribution of labeled vesicles was not significantly different between early (2 min) and late (180 min) time points. Terminals fixed 30 s following stimulation contained a significant proportion of vesicular structures equivalent in diameter to 2-5 regular vesicles, with multivesicular bodies and calveoli rarely seen, suggesting that endocytosis during sustained release at crayfish terminals occurs via multiple routes, most commonly through large "vesicle" intermediates.
Collapse
Affiliation(s)
- Jessica Bilkey
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Patrick C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kerry R Delaney
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
21
|
Yamashita M, Kawaguchi SY, Hori T, Takahashi T. Vesicular GABA Uptake Can Be Rate Limiting for Recovery of IPSCs from Synaptic Depression. Cell Rep 2019; 22:3134-3141. [PMID: 29562170 DOI: 10.1016/j.celrep.2018.02.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/16/2022] Open
Abstract
Synaptic efficacy plays crucial roles in neuronal circuit operation and synaptic plasticity. Presynaptic determinants of synaptic efficacy are neurotransmitter content in synaptic vesicles and the number of vesicles undergoing exocytosis at a time. Bursts of presynaptic firings depress synaptic efficacy, mainly due to depletion of releasable vesicles, whereas recovery from strong depression is initiated by endocytic vesicle retrieval followed by refilling of vesicles with neurotransmitter. We washed out presynaptic cytosolic GABA to induce a rundown of IPSCs at cerebellar inhibitory cell pairs in slices from rats and then allowed fast recovery by elevating GABA concentration using photo-uncaging. The time course of this recovery coincided with that of IPSCs from activity-dependent depression induced by a train of high-frequency stimulation. We conclude that vesicular GABA uptake can be a limiting step for the recovery of inhibitory neurotransmission from synaptic depression.
Collapse
Affiliation(s)
- Manami Yamashita
- Laboratory of Molecular Synaptic Function, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan; Department of Physiology, Faculty of Medicine, Osaka Medical College, Osaka 569-8686, Japan
| | - Shin-Ya Kawaguchi
- Society-Academia Collaboration for Innovation, Kyoto University, Kyoto 606-8501, Japan
| | - Tetsuya Hori
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology (OIST) Graduate University, Okinawa 904-0495, Japan.
| |
Collapse
|
22
|
Upreti C, Konstantinov E, Kassabov SR, Bailey CH, Kandel ER. Serotonin Induces Structural Plasticity of Both Extrinsic Modulating and Intrinsic Mediating Circuits In Vitro in Aplysia Californica. Cell Rep 2019; 28:2955-2965.e3. [PMID: 31509754 DOI: 10.1016/j.celrep.2019.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/27/2018] [Accepted: 07/31/2019] [Indexed: 01/29/2023] Open
Abstract
Long-term sensitization of the gill withdrawal reflex in Aplysia requires heterosynaptic, modulatory input that is mediated in part by the growth of new synaptic connections between sensory neurons and their follower cells (intrinsic mediating circuit). Whether modulatory interneurons (the extrinsic modulatory circuit) also display learning-related structural synaptic plasticity remains unknown. To test this idea, we added a bona fide serotonergic modulatory neuron, the metacerebral cell (MCC), to sensory-motor neuron co-cultures and examined the modulating presynaptic varicosities of MCCs before and after repeated pulses of serotonin (5-HT) that induced long-term facilitation (LTF). We observed robust growth of new serotonergic varicosities that were positive for serotonin and capable of synaptic recycling. Our findings demonstrate that, in addition to structural changes in the intrinsic mediating circuit, there are also significant learning-related structural changes in the extrinsic modulating circuit, and these changes might provide a cellular mechanism for savings and for spread of memory.
Collapse
Affiliation(s)
- Chirag Upreti
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | | | - Stefan R Kassabov
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Craig H Bailey
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Kavli Institute for Brain Science, New York, NY 10027, USA
| | - Eric R Kandel
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Kavli Institute for Brain Science, New York, NY 10027, USA; Howard Hughes Medical Institute, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
23
|
Li S, You M, Chai W, Xu Y, Wang Y. Developmental exposure to nonylphenol induced rat axonal injury in vivo and in vitro. Arch Toxicol 2019; 93:2673-2687. [PMID: 31456014 DOI: 10.1007/s00204-019-02536-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/14/2019] [Indexed: 01/20/2023]
Abstract
Increasing evidence indicates that developmental exposure to nonylphenol (NP) causes damage to the central nervous system (CNS). As the most unique and primary component of neuron, axon is an essential structure for the CNS function. Here, we investigated whether developmental exposure to NP affected rat axonal development in vivo and in vitro. Our results showed that developmental exposure to NP 10, 50, and 100 mg/(kg day) caused an obvious decrease in axonal length and density in the hippocampus. Developmental exposure to NP also altered the expression of CRMP-2 and p-CRMP-2, and activated Wnt-Dvl-GSK-3β cascade in the hippocampus, the crucial signaling that regulates axonal development. Even months after the exposure, impairment of axonal growth and alteration of this cascade were not fully restored. In the primary cultured neurons, 30, 50, and 70 μM NP treatment decreased axonal length and impaired axonal function. Similar to in vivo results, it also activated Wnt-Dvl-GSK-3β cascade in cultured neurons. SB-216763, a specific GSK-3β inhibitor, recovered the shortening of axon and the impairment of axonal function induced by NP. Taken together, our results support the idea that exposure to NP induces axonal injury in the developing neurons. Furthermore, the activation of Wnt-Dvl-GSK-3β cascade contributes to the axonal injury induced by NP.
Collapse
Affiliation(s)
- Siyao Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, Liaoning, People's Republic of China
| | - Mingdan You
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, Liaoning, People's Republic of China
| | - Wenjie Chai
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, Liaoning, People's Republic of China
| | - Yuanyuan Xu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, Liaoning, People's Republic of China.
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
24
|
Koyanagi Y, Torturo CL, Cook DC, Zhou Z, Hemmings HC. Role of specific presynaptic calcium channel subtypes in isoflurane inhibition of synaptic vesicle exocytosis in rat hippocampal neurones. Br J Anaesth 2019; 123:219-227. [PMID: 31056238 PMCID: PMC6676046 DOI: 10.1016/j.bja.2019.03.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/24/2019] [Accepted: 03/16/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND P/Q- and N-type voltage-gated calcium channels (VGCC) are the principal subtypes mediating synaptic vesicle (SV) exocytosis. Both the degree of isoflurane inhibition of SV exocytosis and VGCC subtype expression vary between brain regions and neurotransmitter phenotype. We hypothesised that differences in VGCC subtype expression contribute to synapse-selective presynaptic effects of isoflurane. METHODS We used quantitative live-cell imaging to measure exocytosis in cultured rat hippocampal neurones after transfection of the fluorescent biosensor vGlut1-pHluorin. Selective inhibitors of P/Q- and N-type VGCCs were used to isolate subtype-specific effects of isoflurane. RESULTS Inhibition of N-type channels by 1 μM ω-conotoxin GVIA reduced SV exocytosis to 81±5% of control (n=10). Residual exocytosis mediated by P/Q-type channels was further inhibited by isoflurane to 42±4% of control (n=10). The P/Q-type channel inhibitor ω-agatoxin IVA at 0.4 μM inhibited SV exocytosis to 29±3% of control (n=10). Residual exocytosis mediated by N-type channels was further inhibited by isoflurane to 17±3% of control (n=10). Analysis of isoflurane effects at the level of individual boutons revealed no difference in sensitivity to isoflurane between P/Q- or N-type channel-mediated SV exocytosis (P=0.35). There was no correlation between the effect of agatoxin (P=0.91) or conotoxin (P=0.15) and the effect of isoflurane on exocytosis. CONCLUSIONS Sensitivity of SV exocytosis to isoflurane in rat hippocampal neurones is independent of the specific VGCC subtype coupled to exocytosis. The differential sensitivity of VGCC subtypes to isoflurane does not explain the observed neurotransmitter-selective effects of isoflurane in hippocampus.
Collapse
Affiliation(s)
- Yuko Koyanagi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Department of Anesthesiology, Nihon University School of Dentistry, Tokyo, Japan
| | | | - Daniel C Cook
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Zhenyu Zhou
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Intracellular Zn 2+ transients modulate global gene expression in dissociated rat hippocampal neurons. Sci Rep 2019; 9:9411. [PMID: 31253848 PMCID: PMC6598991 DOI: 10.1038/s41598-019-45844-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/07/2019] [Indexed: 12/22/2022] Open
Abstract
Zinc (Zn2+) is an integral component of many proteins and has been shown to act in a regulatory capacity in different mammalian systems, including as a neurotransmitter in neurons throughout the brain. While Zn2+ plays an important role in modulating neuronal potentiation and synaptic plasticity, little is known about the signaling mechanisms of this regulation. In dissociated rat hippocampal neuron cultures, we used fluorescent Zn2+ sensors to rigorously define resting Zn2+ levels and stimulation-dependent intracellular Zn2+ dynamics, and we performed RNA-Seq to characterize Zn2+-dependent transcriptional effects upon stimulation. We found that relatively small changes in cytosolic Zn2+ during stimulation altered expression levels of 931 genes, and these Zn2+ dynamics induced transcription of many genes implicated in neurite expansion and synaptic growth. Additionally, while we were unable to verify the presence of synaptic Zn2+ in these cultures, we did detect the synaptic vesicle Zn2+ transporter ZnT3 and found it to be substantially upregulated by cytosolic Zn2+ increases. These results provide the first global sequencing-based examination of Zn2+-dependent changes in transcription and identify genes that may mediate Zn2+-dependent processes and functions.
Collapse
|
26
|
Jin Y, Seo KH, Ko HM, Jung TW, Chung YH, Lee JH, Park HH, Kim HC, Jeong JH, Lee SH. Various approaches for measurement of synaptic vesicle endocytosis at the central nerve terminal. Arch Pharm Res 2019; 42:455-465. [PMID: 31115782 DOI: 10.1007/s12272-019-01161-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/16/2019] [Indexed: 10/26/2022]
Abstract
At the presynaptic terminal, neurotransmitters are stored in synaptic vesicles (SVs), which are released and recycled via exo- and endocytosis. SV endocytosis is crucial for sustaining synaptic transmission by maintaining the SV pool. Many studies have shown that presynaptic dysfunction, particularly impairment of SV endocytosis, is related to neurological disorders. Notably, the presynaptic terminal is considered to be a sensitive structure because certain presynaptic dysfunctions, manifested as impaired SV endocytosis or ultrastructural changes in the presynaptic terminal, can be observed before there is a biochemical or pathological evidence of a neurological disorder. Therefore, monitoring and assessing the presynaptic function by SV endocytosis facilitates the development of early markers for neurological disorders. In this study, we reviewed the current methods for assessing and visualizing SV endocytosis at the central nerve terminal.
Collapse
Affiliation(s)
- Yeonsun Jin
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kyoung Hee Seo
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hyun Myung Ko
- Department of Life Science, College of Science and Technology, Woosuk University, Jincheon, 27841, Republic of Korea
| | - Tae Woo Jung
- Research Administration Team, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jong Hyuk Lee
- Department of Pharmaceutical Engineering, College of Life and Health Science, Hoseo University, Asan, 31499, Republic of Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
27
|
Sanderson TM, Bradley CA, Georgiou J, Hong YH, Ng AN, Lee Y, Kim HD, Kim D, Amici M, Son GH, Zhuo M, Kim K, Kaang BK, Kim SJ, Collingridge GL. The Probability of Neurotransmitter Release Governs AMPA Receptor Trafficking via Activity-Dependent Regulation of mGluR1 Surface Expression. Cell Rep 2018; 25:3631-3646.e3. [PMID: 30590038 PMCID: PMC6315206 DOI: 10.1016/j.celrep.2018.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 09/28/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
A major mechanism contributing to synaptic plasticity involves alterations in the number of AMPA receptors (AMPARs) expressed at synapses. Hippocampal CA1 synapses, where this process has been most extensively studied, are highly heterogeneous with respect to their probability of neurotransmitter release, P(r). It is unknown whether there is any relationship between the extent of plasticity-related AMPAR trafficking and the initial P(r) of a synapse. To address this question, we induced metabotropic glutamate receptor (mGluR) dependent long-term depression (mGluR-LTD) and assessed AMPAR trafficking and P(r) at individual synapses, using SEP-GluA2 and FM4-64, respectively. We found that either pharmacological or synaptic activation of mGluR1 reduced synaptic SEP-GluA2 in a manner that depends upon P(r); this process involved an activity-dependent reduction in surface mGluR1 that selectively protects high-P(r) synapses from synaptic weakening. Consequently, the extent of postsynaptic plasticity can be pre-tuned by presynaptic activity.
Collapse
Affiliation(s)
- Thomas M Sanderson
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; Neuroscience Research Institute, Seoul National University College of Medicine, 28 Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea; School of Physiology, Pharmacology & Neuroscience, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Clarrisa A Bradley
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; Neuroscience & Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Yun Hwa Hong
- Neuroscience Research Institute, Seoul National University College of Medicine, 28 Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea; Department of Physiology, Seoul National University College of Medicine, 28, Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea
| | - Ai Na Ng
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Yeseul Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; School of Physiology, Pharmacology & Neuroscience, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Hee-Dae Kim
- Department of Brain and Cognitive Sciences, DGIST, and Korea Brain Institute (KBRI), Daegu, 41068, Korea
| | - Doyeon Kim
- Department of Brain and Cognitive Sciences, DGIST, and Korea Brain Institute (KBRI), Daegu, 41068, Korea
| | - Mascia Amici
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Gi Hoon Son
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 136-705, Seoul, Korea
| | - Min Zhuo
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biological Sciences, College of Natural Sciences, Seoul National University, Building 504, Room 202, 599 Gwanangno, Gwanak-gu 151-747, Seoul, Korea; Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Kyungjin Kim
- Department of Brain and Cognitive Sciences, DGIST, and Korea Brain Institute (KBRI), Daegu, 41068, Korea
| | - Bong-Kiun Kaang
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; Department of Biological Sciences, College of Natural Sciences, Seoul National University, Building 504, Room 202, 599 Gwanangno, Gwanak-gu 151-747, Seoul, Korea; Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Sang Jeong Kim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; Neuroscience Research Institute, Seoul National University College of Medicine, 28 Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea; Department of Physiology, Seoul National University College of Medicine, 28, Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea.
| | - Graham L Collingridge
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 151-746, Korea; School of Physiology, Pharmacology & Neuroscience, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
28
|
Chen S, Yu C, Rong L, Li CH, Qin X, Ryu H, Park H. Altered Synaptic Vesicle Release and Ca 2+ Influx at Single Presynaptic Terminals of Cortical Neurons in a Knock-in Mouse Model of Huntington's Disease. Front Mol Neurosci 2018; 11:478. [PMID: 30618623 PMCID: PMC6311661 DOI: 10.3389/fnmol.2018.00478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the abnormal expansion of CAG repeats in the huntingtin (HTT) gene, which leads to progressive loss of neurons starting in the striatum and cortex. One possible mechanism for this selective loss of neurons in the early stage of HD is altered neurotransmission at synapses. Despite the recent finding that presynaptic terminals play an important role in HD, neurotransmitter release at synapses in HD remains poorly understood. Here, we measured synaptic vesicle release in real time at single presynaptic terminals during electrical field stimulation. We found the increase in synaptic vesicle release at presynaptic terminals in primary cortical neurons in a knock-in mouse model of HD (zQ175). We also found the increase in Ca2+ influx at presynaptic terminals in HD neurons during the electrical stimulation. Consistent with increased Ca2+-dependent neurotransmission in HD neurons, the increase in vesicle release and Ca2+ influx was rescued with Ca2+ chelators or by blocking N-type voltage-gated Ca2+ channels, suggesting N-type voltage-gated Ca2+ channels play an important role in HD. Taken together, our results suggest that the increased synaptic vesicles release due to increased Ca2+ influx at presynaptic terminals in cortical neurons contributes to the selective neurodegeneration of these neurons in early HD and provide a possible therapeutic target.
Collapse
Affiliation(s)
- Sidong Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Chenglong Yu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Li Rong
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Chun Hei Li
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Xianan Qin
- Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Hoon Ryu
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Hyokeun Park
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong.,Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| |
Collapse
|
29
|
Koch ET, Woodard CL, Raymond LA. Direct assessment of presynaptic modulation of cortico-striatal glutamate release in a Huntington’s disease mouse model. J Neurophysiol 2018; 120:3077-3084. [DOI: 10.1152/jn.00638.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the brain, and impairments in its signaling are associated with many neurological disorders, including Huntington’s disease (HD). Previous studies in HD mouse models demonstrate altered glutamate receptor distribution and signaling at cortico-striatal synapses, and some studies suggest that glutamate release is altered; however, traditional methods to study synaptic glutamate release are indirect or have poor temporal resolution. Here we utilize iGluSnFR, a modified green fluorescent protein reporter for real-time imaging of glutamate transmission, to study presynaptic modulation of cortical glutamate release in the striatum of the YAC128 HD mouse model. We determined that iGluSnFR can be used to accurately measure short- and long-term changes in glutamate release caused by modulation of extracellular Ca2+ levels, activation of presynaptic receptors, and high-frequency stimulation (HFS) protocols. We also confirmed a difference in the expression of HFS-induced long-term depression in YAC128. Together, this research demonstrates the utility of iGluSnFR in studying presynaptic modulation of glutamate release in healthy mice and disease models that display impairments in glutamate signaling. NEW & NOTEWORTHY We use iGluSnFR to directly assess presynaptic modulation of cortico-striatal glutamate release in brain slice and compare changes in glutamate release between wild type and a Huntington’s disease mouse model, YAC128. We observed reductions in glutamate release after low extracellular Ca2+ and activation of various presynaptic receptors. We also demonstrate a presynaptic mechanism of reduced glutamate release in high-frequency stimulation-induced long-term depression and show this to be altered in YAC128.
Collapse
Affiliation(s)
- Ellen T. Koch
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cameron L. Woodard
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lynn A. Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Song Q, Huang M, Wang B, Kang X, Wang C. Bidirectional regulation of Ca 2+ in exo-endocytosis coupling. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1583-1585. [PMID: 30484064 DOI: 10.1007/s11427-018-9429-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Qian Song
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Mingzhu Huang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.,College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Bianbian Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.,College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Xinjiang Kang
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.,DOE-Key Lab of Medical Electrophysiology, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Changhe Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
31
|
Midorikawa M. Real-time imaging of synaptic vesicle exocytosis by total internal reflection fluorescence (TIRF) microscopy. Neurosci Res 2018; 136:1-5. [DOI: 10.1016/j.neures.2018.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/29/2018] [Indexed: 12/15/2022]
|
32
|
Gan Q, Watanabe S. Synaptic Vesicle Endocytosis in Different Model Systems. Front Cell Neurosci 2018; 12:171. [PMID: 30002619 PMCID: PMC6031744 DOI: 10.3389/fncel.2018.00171] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/01/2018] [Indexed: 11/13/2022] Open
Abstract
Neurotransmission in complex animals depends on a choir of functionally distinct synapses releasing neurotransmitters in a highly coordinated manner. During synaptic signaling, vesicles fuse with the plasma membrane to release their contents. The rate of vesicle fusion is high and can exceed the rate at which synaptic vesicles can be re-supplied by distant sources. Thus, local compensatory endocytosis is needed to replenish the synaptic vesicle pools. Over the last four decades, various experimental methods and model systems have been used to study the cellular and molecular mechanisms underlying synaptic vesicle cycle. Clathrin-mediated endocytosis is thought to be the predominant mechanism for synaptic vesicle recycling. However, recent studies suggest significant contribution from other modes of endocytosis, including fast compensatory endocytosis, activity-dependent bulk endocytosis, ultrafast endocytosis, as well as kiss-and-run. Currently, it is not clear whether a universal model of vesicle recycling exist for all types of synapses. It is possible that each synapse type employs a particular mode of endocytosis. Alternatively, multiple modes of endocytosis operate at the same synapse, and the synapse toggles between different modes depending on its activity level. Here we compile review and research articles based on well-characterized model systems: frog neuromuscular junctions, C. elegans neuromuscular junctions, Drosophila neuromuscular junctions, lamprey reticulospinal giant axons, goldfish retinal ribbon synapses, the calyx of Held, and rodent hippocampal synapses. We will compare these systems in terms of their known modes and kinetics of synaptic vesicle endocytosis, as well as the underlying molecular machineries. We will also provide the future development of this field.
Collapse
Affiliation(s)
- Quan Gan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
33
|
Kopke DL, Broadie K. FM Dye Cycling at the Synapse: Comparing High Potassium Depolarization, Electrical and Channelrhodopsin Stimulation. J Vis Exp 2018:57765. [PMID: 29889207 PMCID: PMC6101380 DOI: 10.3791/57765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
FM dyes are used to study the synaptic vesicle (SV) cycle. These amphipathic probes have a hydrophilic head and hydrophobic tail, making them water-soluble with the ability to reversibly enter and exit membrane lipid bilayers. These styryl dyes are relatively non-fluorescent in aqueous medium, but insertion into the outer leaflet of the plasma membrane causes a >40X increase in fluorescence. In neuronal synapses, FM dyes are internalized during SV endocytosis, trafficked both within and between SV pools, and released with SV exocytosis, providing a powerful tool to visualize presynaptic stages of neurotransmission. A primary genetic model of glutamatergic synapse development and function is the Drosophila neuromuscular junction (NMJ), where FM dye imaging has been used extensively to quantify SV dynamics in a wide range of mutant conditions. The NMJ synaptic terminal is easily accessible, with a beautiful array of large synaptic boutons ideal for imaging applications. Here, we compare and contrast the three ways to stimulate the Drosophila NMJ to drive activity-dependent FM1-43 dye uptake/release: 1) bath application of high [K+] to depolarize neuromuscular tissues, 2) suction electrode motor nerve stimulation to depolarize the presynaptic nerve terminal, and 3) targeted transgenic expression of channelrhodopsin variants for light-stimulated, spatial control of depolarization. Each of these methods has benefits and disadvantages for the study of genetic mutation effects on the SV cycle at the Drosophila NMJ. We will discuss these advantages and disadvantages to assist the selection of the stimulation approach, together with the methodologies specific to each strategy. In addition to fluorescent imaging, FM dyes can be photoconverted to electron-dense signals visualized using transmission electron microscopy (TEM) to study SV cycle mechanisms at an ultrastructural level. We provide the comparisons of confocal and electron microscopy imaging from the different methods of Drosophila NMJ stimulation, to help guide the selection of future experimental paradigms.
Collapse
Affiliation(s)
| | - Kendal Broadie
- Departments of Biological Sciences, Pharmacology, Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center;
| |
Collapse
|
34
|
Higashi T, Tanaka S, Iida T, Okabe S. Synapse Elimination Triggered by BMP4 Exocytosis and Presynaptic BMP Receptor Activation. Cell Rep 2018; 22:919-929. [DOI: 10.1016/j.celrep.2017.12.101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/03/2017] [Accepted: 12/26/2017] [Indexed: 11/16/2022] Open
|
35
|
Cousin MA, Gordon SL, Smillie KJ. Using FM Dyes to Monitor Clathrin-Mediated Endocytosis in Primary Neuronal Culture. Methods Mol Biol 2018; 1847:239-249. [PMID: 30129022 DOI: 10.1007/978-1-4939-8719-1_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This protocol utilizes lipophilic FM dyes to monitor membrane recycling in real time. FM dyes are virtually nonfluorescent in solution but when membrane bound are intensely fluorescent, combined with the flexibility of different emission wavelengths make these dyes an excellent choice for investigating clathrin-mediated endocytosis, among other membrane trafficking and recycling pathways.
Collapse
Affiliation(s)
- Michael A Cousin
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Sarah L Gordon
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Karen J Smillie
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
36
|
Rampérez A, Sánchez-Prieto J, Torres M. Brefeldin A sensitive mechanisms contribute to endocytotic membrane retrieval and vesicle recycling in cerebellar granule cells. J Neurochem 2017; 141:662-675. [PMID: 28295320 DOI: 10.1111/jnc.14017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 11/26/2022]
Abstract
The recycling of synaptic vesicle (SV) proteins and transmitter release occur at multiple sites along the axon. These processes are sensitive to inhibition of the small GTP binding protein ARF1, which regulates the adaptor protein 1 and 3 complex (AP-1/AP-3). As the axon matures, SV recycling becomes restricted to the presynaptic bouton, and its machinery undergoes a complex process of maturation. We used the styryl dye FM1-43 to highlight differences in the efficiency of membrane recycling at different sites in cerebellar granule cells cultured for 7 days in vitro. We used Brefeldin A (BFA) to inhibit AP-1/AP-3-mediated recycling and to test the contribution of this pathway to the heterogeneity of the responses when these cells are strongly stimulated. Combining imaging techniques and ultrastructural analyses, we found a significant decrease in the density of functional boutons and an increase in the presence of endosome-like structures within the boutons of cells incubated with BFA prior to FM1-43 loading. Such effects were not observed when BFA was added 5 min after the end of the loading step, when endocytosis was almost fully completed. In this situation, vesicles were found closer to the active zone (AZ) in boutons exposed to BFA. Together, these data suggest that the AP-1/AP-3 pathway contributes to SV recycling, affecting different steps in all boutons but not equally, and thus being partly responsible for the heterogeneity of the different recycling efficiencies. Cover Image for this issue: doi. 10.1111/jnc.13801.
Collapse
Affiliation(s)
- Alberto Rampérez
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Magdalena Torres
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
37
|
Xie Z, Long J, Liu J, Chai Z, Kang X, Wang C. Molecular Mechanisms for the Coupling of Endocytosis to Exocytosis in Neurons. Front Mol Neurosci 2017; 10:47. [PMID: 28348516 PMCID: PMC5346583 DOI: 10.3389/fnmol.2017.00047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/10/2017] [Indexed: 11/13/2022] Open
Abstract
Neuronal communication and brain function mainly depend on the fundamental biological events of neurotransmission, including the exocytosis of presynaptic vesicles (SVs) for neurotransmitter release and the subsequent endocytosis for SV retrieval. Neurotransmitters are released through the Ca2+- and SNARE-dependent fusion of SVs with the presynaptic plasma membrane. Following exocytosis, endocytosis occurs immediately to retrieve SV membrane and fusion machinery for local recycling and thus maintain the homeostasis of synaptic structure and sustained neurotransmission. Apart from the general endocytic machinery, recent studies have also revealed the involvement of SNARE proteins (synaptobrevin, SNAP25 and syntaxin), synaptophysin, Ca2+/calmodulin, and members of the synaptotagmin protein family (Syt1, Syt4, Syt7 and Syt11) in the balance and tight coupling of exo-endocytosis in neurons. Here, we provide an overview of recent progress in understanding how these neuron-specific adaptors coordinate to ensure precise and efficient endocytosis during neurotransmission.
Collapse
Affiliation(s)
- Zhenli Xie
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong UniversityXi'an, China; Frontier Institute of Science and Technology, Xi'an Jiaotong UniversityXi'an, China; State Key Laboratory of Membrane Biology, Peking UniversityBeijing, China; Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking UniversityBeijing, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong UniversityXi'an, China; Frontier Institute of Science and Technology, Xi'an Jiaotong UniversityXi'an, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong UniversityXi'an, China; Frontier Institute of Science and Technology, Xi'an Jiaotong UniversityXi'an, China
| | - Zuying Chai
- State Key Laboratory of Membrane Biology, Peking UniversityBeijing, China; Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking UniversityBeijing, China
| | - Xinjiang Kang
- State Key Laboratory of Membrane Biology, Peking UniversityBeijing, China; Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking UniversityBeijing, China; College of Life Sciences, Liaocheng UniversityLiaocheng, China; Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical UniversityLuzhou, China
| | - Changhe Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong UniversityXi'an, China; Frontier Institute of Science and Technology, Xi'an Jiaotong UniversityXi'an, China; State Key Laboratory of Membrane Biology, Peking UniversityBeijing, China; Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking UniversityBeijing, China
| |
Collapse
|
38
|
Joensuu M, Padmanabhan P, Durisic N, Bademosi ATD, Cooper-Williams E, Morrow IC, Harper CB, Jung W, Parton RG, Goodhill GJ, Papadopulos A, Meunier FA. Subdiffractional tracking of internalized molecules reveals heterogeneous motion states of synaptic vesicles. J Cell Biol 2016; 215:277-292. [PMID: 27810917 PMCID: PMC5080683 DOI: 10.1083/jcb.201604001] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/30/2016] [Indexed: 11/23/2022] Open
Abstract
Joensuu et al. describe a tool for subdiffractional tracking of internalized molecules. They reveal that synaptic vesicles exhibit stochastic switching between heterogeneous diffusive and transport states in live hippocampal nerve terminals. Our understanding of endocytic pathway dynamics is severely restricted by the diffraction limit of light microscopy. To address this, we implemented a novel technique based on the subdiffractional tracking of internalized molecules (sdTIM). This allowed us to image anti–green fluorescent protein Atto647N-tagged nanobodies trapped in synaptic vesicles (SVs) from live hippocampal nerve terminals expressing vesicle-associated membrane protein 2 (VAMP2)–pHluorin with 36-nm localization precision. Our results showed that, once internalized, VAMP2–pHluorin/Atto647N–tagged nanobodies exhibited a markedly lower mobility than on the plasma membrane, an effect that was reversed upon restimulation in presynapses but not in neighboring axons. Using Bayesian model selection applied to hidden Markov modeling, we found that SVs oscillated between diffusive states or a combination of diffusive and transport states with opposite directionality. Importantly, SVs exhibiting diffusive motion were relatively less likely to switch to the transport motion. These results highlight the potential of the sdTIM technique to provide new insights into the dynamics of endocytic pathways in a wide variety of cellular settings.
Collapse
Affiliation(s)
- Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nela Durisic
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Adekunle T D Bademosi
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Isabel C Morrow
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Callista B Harper
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - WooRam Jung
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Geoffrey J Goodhill
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.,School of Mathematics and Physics, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andreas Papadopulos
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia .,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia .,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
39
|
Unique pH dynamics in GABAergic synaptic vesicles illuminates the mechanism and kinetics of GABA loading. Proc Natl Acad Sci U S A 2016; 113:10702-7. [PMID: 27601664 DOI: 10.1073/pnas.1604527113] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
GABA acts as the major inhibitory neurotransmitter in the mammalian brain, shaping neuronal and circuit activity. For sustained synaptic transmission, synaptic vesicles (SVs) are required to be recycled and refilled with neurotransmitters using an H(+) electrochemical gradient. However, neither the mechanism underlying vesicular GABA uptake nor the kinetics of GABA loading in living neurons have been fully elucidated. To characterize the process of GABA uptake into SVs in functional synapses, we monitored luminal pH of GABAergic SVs separately from that of excitatory glutamatergic SVs in cultured hippocampal neurons. By using a pH sensor optimal for the SV lumen, we found that GABAergic SVs exhibited an unexpectedly higher resting pH (∼6.4) than glutamatergic SVs (pH ∼5.8). Moreover, unlike glutamatergic SVs, GABAergic SVs displayed unique pH dynamics after endocytosis that involved initial overacidification and subsequent alkalization that restored their resting pH. GABAergic SVs that lacked the vesicular GABA transporter (VGAT) did not show the pH overshoot and acidified further to ∼6.0. Comparison of luminal pH dynamics in the presence or absence of VGAT showed that VGAT operates as a GABA/H(+) exchanger, which is continuously required to offset GABA leakage. Furthermore, the kinetics of GABA transport was slower (τ > 20 s at physiological temperature) than that of glutamate uptake and may exceed the time required for reuse of exocytosed SVs, allowing reuse of incompletely filled vesicles in the presence of high demand for inhibitory transmission.
Collapse
|
40
|
Endosome-mediated endocytic mechanism replenishes the majority of synaptic vesicles at mature CNS synapses in an activity-dependent manner. Sci Rep 2016; 6:31807. [PMID: 27534442 PMCID: PMC4989163 DOI: 10.1038/srep31807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/26/2016] [Indexed: 11/30/2022] Open
Abstract
Whether synaptic vesicles (SVs) are recovered via endosome-mediated pathways is a matter of debate; however, recent evidence suggests that clathrin-independent bulk endocytosis (CIE) via endosomes is functional and preferentially replenishes SV pools during strong stimulation. Here, using brefeldin-A (BFA) to block CIE, we found that CIE retrieved a minority of SVs at developing CNS synapses during strong stimulation, but its contribution increased up to 61% at mature CNS synapses. Contrary to previous views, BFA not only blocked SV formation from the endosome but also blocked the endosome formation at the plasma membrane. Adaptor protein 1 and 3 (AP-1/3) have key roles in SV reformation from endosomes during CIE, and AP-1 also affects bulk endosome formation from the plasma membrane. Finally, temporary blocking of chronic or acute neuronal activity with tetrodotoxin in mature neurons redirected most SV retrieval to endosome-independent pathways. These results show that during high neuronal activity, CIE becomes the major endocytic pathway at mature CNS synapses. Moreover, mature neurons use clathrin-mediated endocytosis and the CIE pathway to different extents depending on their previous activity; this may result in activity-dependent alterations of the SV composition which ultimately influence transmitter release and contribute to synaptic plasticity.
Collapse
|
41
|
Marshall KE, Vadukul DM, Dahal L, Theisen A, Fowler MW, Al-Hilaly Y, Ford L, Kemenes G, Day IJ, Staras K, Serpell LC. A critical role for the self-assembly of Amyloid-β1-42 in neurodegeneration. Sci Rep 2016; 6:30182. [PMID: 27443509 PMCID: PMC4957119 DOI: 10.1038/srep30182] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 06/28/2016] [Indexed: 11/23/2022] Open
Abstract
Amyloid β1-42 (Aβ1-42) plays a central role in Alzheimer’s disease. The link between structure, assembly and neuronal toxicity of this peptide is of major current interest but still poorly defined. Here, we explored this relationship by rationally designing a variant form of Aβ1-42 (vAβ1-42) differing in only two amino acids. Unlike Aβ1-42, we found that the variant does not self-assemble, nor is it toxic to neuronal cells. Moreover, while Aβ1-42 oligomers impact on synaptic function, vAβ1-42 does not. In a living animal model system we demonstrate that only Aβ1-42 leads to memory deficits. Our findings underline a key role for peptide sequence in the ability to assemble and form toxic structures. Furthermore, our non-toxic variant satisfies an unmet demand for a closely related control peptide for Aβ1-42 cellular studies of disease pathology, offering a new opportunity to decipher the mechanisms that accompany Aβ1-42-induced toxicity leading to neurodegeneration.
Collapse
Affiliation(s)
- Karen E Marshall
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Devkee M Vadukul
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Liza Dahal
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Alina Theisen
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Milena W Fowler
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Youssra Al-Hilaly
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK.,College of Sciences, Chemistry Department, Al-Mustansiriyah University, Baghdad, Iraq
| | - Lenzie Ford
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - György Kemenes
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Iain J Day
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Kevin Staras
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| | - Louise C Serpell
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG, UK
| |
Collapse
|
42
|
Plattner H. Trichocysts-Paramecium'sProjectile-like Secretory Organelles. J Eukaryot Microbiol 2016; 64:106-133. [DOI: 10.1111/jeu.12332] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/09/2016] [Accepted: 05/21/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Helmut Plattner
- Department of Biology; University of Konstanz; PO Box M625 78457 Konstanz Germany
| |
Collapse
|
43
|
Sun Q, Weinger JG, Mao F, Liu G. Regulation of structural and functional synapse density by L-threonate through modulation of intraneuronal magnesium concentration. Neuropharmacology 2016; 108:426-39. [PMID: 27178134 DOI: 10.1016/j.neuropharm.2016.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/18/2016] [Accepted: 05/09/2016] [Indexed: 12/25/2022]
Abstract
Oral administration of the combination of L-threonate (threonate) and magnesium (Mg(2+)) in the form of L-Threonic acid Magnesium salt (L-TAMS) can enhance learning and memory in young rats and prevent memory decline in aging rats and in Alzheimer's disease model mice. Recent results from a human clinical trial demonstrate the efficacy of L-TAMS in restoring global cognitive abilities of older adults. Previously, we reported that neuronal intracellular Mg(2+) serves as a critical signaling molecule for controlling synapse density, a key factor that determines cognitive ability. The elevation of brain Mg(2+) by oral administration of L-TAMS in intact animals plays a significant role in mediating the therapeutic effects of L-TAMS. The current study sought to elucidate the unique role of threonate. We aimed to understand if threonate acts directly to elevate intraneuronal Mg(2+), and why Mg(2+) given without threonate is ineffective for enhancing learning and memory ability. We discovered that threonate is naturally present in cerebrospinal fluid (CSF) and oral treatment with L-TAMS elevated CSF threonate. In cultured hippocampal neurons, threonate treatment directly induced an increase in intracellular Mg(2+) concentration. Functionally, elevating threonate upregulated expression of NR2B-containing NMDAR, boosted mitochondrial membrane potential (ΔΨm), and increased functional synapse density in neuronal cultures. These effects are unique to threonate, as other common Mg(2+) anions failed to have the same results. Mechanistically, threonate's effects were specifically mediated through glucose transporters (GLUTs). We also evaluated the effects of threonate in human neural stem cell-derived neurons, and found it was equally effective at upregulating synapse density. The current study provides an explanation for why threonate is an essential component of L-TAMS and supports the use of L-TAMS to promote cognitive abilities in human.
Collapse
Affiliation(s)
- Qifeng Sun
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | | | - Fei Mao
- Neurocentria, Inc., Fremont, CA 94538, USA
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China; Neurocentria, Inc., Fremont, CA 94538, USA.
| |
Collapse
|
44
|
Rauti R, Lozano N, León V, Scaini D, Musto M, Rago I, Ulloa Severino FP, Fabbro A, Casalis L, Vázquez E, Kostarelos K, Prato M, Ballerini L. Graphene Oxide Nanosheets Reshape Synaptic Function in Cultured Brain Networks. ACS NANO 2016; 10:4459-71. [PMID: 27030936 DOI: 10.1021/acsnano.6b00130] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Graphene offers promising advantages for biomedical applications. However, adoption of graphene technology in biomedicine also poses important challenges in terms of understanding cell responses, cellular uptake, or the intracellular fate of soluble graphene derivatives. In the biological microenvironment, graphene nanosheets might interact with exposed cellular and subcellular structures, resulting in unexpected regulation of sophisticated biological signaling. More broadly, biomedical devices based on the design of these 2D planar nanostructures for interventions in the central nervous system require an accurate understanding of their interactions with the neuronal milieu. Here, we describe the ability of graphene oxide nanosheets to down-regulate neuronal signaling without affecting cell viability.
Collapse
Affiliation(s)
- Rossana Rauti
- Life Science Department, University of Trieste , 34127 Trieste, Italy
| | - Neus Lozano
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Veronica León
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Denis Scaini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | - Mattia Musto
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| | - Ilaria Rago
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | | | - Alessandra Fabbro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
| | | | - Ester Vázquez
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Kostas Kostarelos
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
- CIC BiomaGUNE, Parque Tecnológico de San Sebastián, Paseo Miramón, 182, 20009 San Sebastián, Guipúzcoa, Spain
- Basque Foundation for Science , Ikerbasque, Bilbao 48013, Spain
| | - Laura Ballerini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| |
Collapse
|
45
|
Nedelescu H, Chowdhury TG, Wable GS, Arbuthnott G, Aoki C. Cerebellar sub-divisions differ in exercise-induced plasticity of noradrenergic axons and in their association with resilience to activity-based anorexia. Brain Struct Funct 2016; 222:317-339. [PMID: 27056728 PMCID: PMC5215061 DOI: 10.1007/s00429-016-1220-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/24/2016] [Indexed: 11/01/2022]
Abstract
The vermis or "spinocerebellum" receives input from the spinal cord and motor cortex for controlling balance and locomotion, while the longitudinal hemisphere region or "cerebro-cerebellum" is interconnected with non-motor cortical regions, including the prefrontal cortex that underlies decision-making. Noradrenaline release in the cerebellum is known to be important for motor plasticity but less is known about plasticity of the cerebellar noradrenergic (NA) system, itself. We characterized plasticity of dopamine β-hydroxylase-immunoreactive NA fibers in the cerebellum of adolescent female rats that are evoked by voluntary wheel running, food restriction (FR) or by both, in combination. When 8 days of wheel access was combined with FR during the last 4 days, some responded with excessive exercise, choosing to run even during the hours of food access: this exacerbated weight loss beyond that due to FR alone. In the vermis, exercise, with or without FR, shortened the inter-varicosity intervals and increased varicosity density along NA fibers, while excessive exercise, due to FR, also shortened NA fibers. In contrast, the hemisphere required the FR-evoked excessive exercise to evoke shortened inter-varicosity intervals along NA fibers and this change was exhibited more strongly by rats that suppressed the FR-evoked excessive exercise, a behavior that minimized weight loss. Presuming that shortened inter-varicosity intervals translate to enhanced NA release and synthesis of norepinephrine, this enhancement in the cerebellar hemisphere may contribute towards protection of individuals from the life-threatening activity-based anorexia via relays with higher-order cortical areas that mediate the animal's decision to suppress the innate FR-evoked hyperactivity.
Collapse
Affiliation(s)
- Hermina Nedelescu
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan. .,Center for Neural Science, New York University, New York, NY, 10003, USA. .,Brain Mechanisms for Behaviour Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan.
| | - Tara G Chowdhury
- Center for Neural Science, New York University, New York, NY, 10003, USA.,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Gauri S Wable
- Center for Neural Science, New York University, New York, NY, 10003, USA
| | - Gordon Arbuthnott
- Brain Mechanisms for Behaviour Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, NY, 10003, USA.
| |
Collapse
|
46
|
Expression of Quinone Reductase-2 in the Cortex Is a Muscarinic Acetylcholine Receptor-Dependent Memory Consolidation Constraint. J Neurosci 2016; 35:15568-81. [PMID: 26609153 DOI: 10.1523/jneurosci.1170-15.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED Learning of novel information, including novel taste, requires activation of neuromodulatory transmission mediated, for example, by the muscarinic acetylcholine receptors (mAChRs) in relevant brain structures. In addition, drugs enhancing the function of mAChRs are used to treat memory impairment and decline. However, the mechanisms underlying these effects are poorly understood. Here, using quantitative RT-PCR in Wistar Hola rats, we found quinone reductase 2 (QR2) to be expressed in the cortex in an mAChR-dependent manner. QR2 mRNA expression in the insular cortex is inversely correlated with mAChR activation both endogenously, after novel taste learning, and exogenously, after pharmacological manipulation of the muscarinic transmission. Moreover, reducing QR2 expression levels through lentiviral shRNA vectors or activity via inhibitors is sufficient to enhance long-term memories. We also show here that, in patients with Alzheimer's disease, QR2 is overexpressed in the cortex. It is suggested that QR2 expression in the cortex is a removable limiting factor of memory formation and thus serves as a new target to enhance cognitive function and delay the onset of neurodegenerative diseases. SIGNIFICANCE STATEMENT We found that: (1) quinone reductase 2 (QR2) expression is a muscarinic-receptor-dependent removable constraint on memory formation in the cortex, (2) reducing QR2 expression or activity in the cortex enhances memory formation, and (3) Alzheimer's disease patients overexpressed QR2. We believe that these results propose a new mechanism by which muscarinic acetylcholine receptors affect cognition and suggest that inhibition of QR2 is a way to enhance cognition in normal and pathological conditions.
Collapse
|
47
|
Eckenstaler R, Lessmann V, Brigadski T. CAPS1 effects on intragranular pH and regulation of BDNF release from secretory granules in hippocampal neurons. J Cell Sci 2016; 129:1378-90. [PMID: 26869227 DOI: 10.1242/jcs.178251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023] Open
Abstract
The secretory protein brain-derived neurotrophic factor (BDNF) is assumed to be a key factor for the induction of synaptic plasticity processes in neurons. However, the molecular mechanisms for activity-dependent release of the protein largely remain elusive. Here, we demonstrate the relevance of the priming factor CAPS1 (also known as CADPS) for the maturation and exocytosis of BDNF-containing secretory granules, as well as for neurotransmitter release from synaptic vesicles. Using live-cell imaging and RNA silencing methods, we show that CAPS1 has a previously unrecognized function in regulating the intragranular pH of BDNF-containing secretory granules. Furthermore, our results demonstrate that acute single-cell knockdown of CAPS1 with unaltered expression in neighboring neurons leads to a strong reduction in the number of fusion-competent secretory granules and to a significant decrease of released BDNF following exocytosis in dendrites of CAPS1-deficient neurons. In addition, our results show a reduction in synaptic vesicle turnover after CAPS1 knockdown without affecting the density of active boutons in hippocampal neurons. Thus, our results reveal new functions of endogenous CAPS1 in the BDNF secretory granule life cycle, thereby representing a new mechanism of neuronal plasticity.
Collapse
Affiliation(s)
- Robert Eckenstaler
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany
| | - Volkmar Lessmann
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany Center of Behavioral Brain Sciences (CBBS), Magdeburg 39120, Germany
| | - Tanja Brigadski
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, Magdeburg 39120, Germany Center of Behavioral Brain Sciences (CBBS), Magdeburg 39120, Germany
| |
Collapse
|
48
|
Rey SA, Smith CA, Fowler MW, Crawford F, Burden JJ, Staras K. Ultrastructural and functional fate of recycled vesicles in hippocampal synapses. Nat Commun 2015; 6:8043. [PMID: 26292808 PMCID: PMC4560786 DOI: 10.1038/ncomms9043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/11/2015] [Indexed: 12/24/2022] Open
Abstract
Efficient recycling of synaptic vesicles is thought to be critical for sustained information transfer at central terminals. However, the specific contribution that retrieved vesicles make to future transmission events remains unclear. Here we exploit fluorescence and time-stamped electron microscopy to track the functional and positional fate of vesicles endocytosed after readily releasable pool (RRP) stimulation in rat hippocampal synapses. We show that most vesicles are recovered near the active zone but subsequently take up random positions in the cluster, without preferential bias for future use. These vesicles non-selectively queue, advancing towards the release site with further stimulation in an actin-dependent manner. Nonetheless, the small subset of vesicles retrieved recently in the stimulus train persist nearer the active zone and exhibit more privileged use in the next RRP. Our findings reveal heterogeneity in vesicle fate based on nanoscale position and timing rules, providing new insights into the origins of future pool constitution.
Collapse
Affiliation(s)
- Stephanie A Rey
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | | | - Milena W Fowler
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Freya Crawford
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Jemima J Burden
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Kevin Staras
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| |
Collapse
|
49
|
Abstract
During synaptic vesicle (SV) recycling, the vacuolar-type H(+) ATPase creates a proton electrochemical gradient (ΔμH(+)) that drives neurotransmitter loading into SVs. Given the low estimates of free luminal protons, it has been envisioned that the influx of a limited number of protons suffices to establish ΔμH(+). Consistent with this, the time constant of SV re-acidification was reported to be <5 s, much faster than glutamate loading (τ of ∼ 15 s) and thus unlikely to be rate limiting for neurotransmitter loading. However, such estimates have relied on pHluorin-based probes that lack sensitivity in the lower luminal pH range. Here, we reexamined re-acidification kinetics using the mOrange2-based probe that should report the SV pH more accurately. In recordings from cultured mouse hippocampal neurons, we found that re-acidification took substantially longer (τ of ∼ 15 s) than estimated previously. In addition, we found that the SV lumen exhibited a large buffering capacity (∼ 57 mm/pH), corresponding to an accumulation of ∼ 1200 protons during re-acidification. Together, our results uncover hitherto unrecognized robust proton influx and storage in SVs that can restrict the rate of neurotransmitter refilling.
Collapse
|
50
|
Schneggenburger R, Rosenmund C. Molecular mechanisms governing Ca2+ regulation of evoked and spontaneous release. Nat Neurosci 2015; 18:935-41. [DOI: 10.1038/nn.4044] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/09/2015] [Indexed: 12/15/2022]
|