1
|
Xu N, Chen SY, Tang AH. Tuning synapse strength by nanocolumn plasticity. Trends Neurosci 2025; 48:200-212. [PMID: 39848836 DOI: 10.1016/j.tins.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025]
Abstract
The precise organization of the complex set of synaptic proteins at the nanometer scale is crucial for synaptic transmission. At the heart of this nanoscale architecture lies the nanocolumn. This aligns presynaptic neurotransmitter release with a high local density of postsynaptic receptor channels, thereby optimizing synaptic strength. Although synapses exhibit diverse protein compositions and nanoscale organizations, the role of structural diversity in the notable differences observed in synaptic physiology remains poorly understood. In this review we examine the current literature on the molecular mechanisms underlying the formation and maintenance of nanocolumns, as well as their role in modulating various aspects of synaptic transmission. We also discuss how the reorganization of nanocolumns contributes to functional dynamics in both synaptic plasticity and pathology.
Collapse
Affiliation(s)
- Na Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Medicine, Anhui University of Science and Technology, Huainan 232001, China.
| | - Si-Yu Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.
| |
Collapse
|
2
|
Chien C, He K, Perry S, Tchitchkan E, Han Y, Li X, Dickman D. Distinct input-specific mechanisms enable presynaptic homeostatic plasticity. SCIENCE ADVANCES 2025; 11:eadr0262. [PMID: 39951523 PMCID: PMC11827636 DOI: 10.1126/sciadv.adr0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Synapses are endowed with the flexibility to change through experience, but must be sufficiently stable to last a lifetime. This tension is illustrated at the Drosophila neuromuscular junction (NMJ), where two motor inputs that differ in structural and functional properties coinnervate most muscles to coordinate locomotion. To stabilize NMJ activity, motor neurons augment neurotransmitter release following diminished postsynaptic glutamate receptor functionality, termed presynaptic homeostatic potentiation (PHP). How these distinct inputs contribute to PHP plasticity remains enigmatic. We have used a botulinum neurotoxin to selectively silence each input and resolve their roles in PHP, demonstrating that PHP is input specific: Chronic (genetic) PHP selectively targets the tonic MN-Ib, where active zone remodeling enhances Ca2+ influx to promote increased glutamate release. In contrast, acute (pharmacological) PHP selectively increases vesicle pools to potentiate phasic MN-Is. Thus, distinct homeostatic modulations in active zone nanoarchitecture, vesicle pools, and Ca2+ influx collaborate to enable input-specific PHP expression.
Collapse
Affiliation(s)
- Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Kaikai He
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Tchitchkan
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
3
|
Bakshinska D, Liu WY, Schultz R, Stowers RS, Hoagland A, Cypranowska C, Stanley C, Younger SH, Newman ZL, Isacoff EY. Synapse-specific catecholaminergic modulation of neuronal glutamate release. Proc Natl Acad Sci U S A 2025; 122:e2420496121. [PMID: 39793084 PMCID: PMC11725921 DOI: 10.1073/pnas.2420496121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/15/2024] [Indexed: 01/12/2025] Open
Abstract
Norepinephrine in vertebrates and its invertebrate analog, octopamine, regulate the activity of neural circuits. We find that, when hungry, Drosophila larvae switch activity in type II octopaminergic motor neurons (MNs) to high-frequency bursts, which coincide with locomotion-driving bursts in type I glutamatergic MNs that converge on the same muscles. Optical quantal analysis across hundreds of synapses simultaneously reveals that octopamine potentiates glutamate release by tonic type Ib MNs, but not phasic type Is MNs, and occurs via the Gq-coupled octopamine receptor (OAMB). OAMB is more abundant in type Ib terminals and acts through diacylglycerol and its target Unc13A, a key component of the glutamate release machinery. Potentiation varies significantly-by up to 1,000%-across synapses of a single Ib axon, with synaptic Unc13A levels determining both release probability and potentiation. We propose that a dual molecular mechanism-an upstream neuromodulator receptor and a downstream transmitter release controller-fine-tunes catecholaminergic modulation so that strong tonic synapses exhibit large potentiation, while weaker tonic and all phasic synapses maintain consistency, yielding a sophisticated regulation of locomotor behavior.
Collapse
Affiliation(s)
- Dariya Bakshinska
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA94720
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - William YuChen Liu
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - Ryan Schultz
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA94720
| | - R. Steven Stowers
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT59717
| | - Adam Hoagland
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - Caroline Cypranowska
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - Cherise Stanley
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - Susan H. Younger
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - Zachary L. Newman
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
| | - Ehud Y. Isacoff
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA94720
- Department of Neuroscience and Molecular & Cell Biology, University of California, Berkeley, CA94720
- Weill Neurohub, University of California Berkeley,Berkeley, CA94720
- Molecular Biophysics and Integrated BioImaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA94720
| |
Collapse
|
4
|
Lützkendorf J, Matkovic-Rachid T, Liu S, Götz T, Gao L, Turrel O, Maglione M, Grieger M, Putignano S, Ramesh N, Ghelani T, Neumann A, Gimber N, Schmoranzer J, Stawrakakis A, Brence B, Baum D, Ludwig K, Heine M, Mielke T, Liu F, Walter AM, Wahl MC, Sigrist SJ. Blobby is a synaptic active zone assembly protein required for memory in Drosophila. Nat Commun 2025; 16:271. [PMID: 39747038 PMCID: PMC11696761 DOI: 10.1038/s41467-024-55382-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
At presynaptic active zones (AZs), scaffold proteins are critical for coordinating synaptic vesicle release and forming essential nanoarchitectures. However, regulatory principles steering AZ scaffold assembly, function, and plasticity remain insufficiently understood. We here identify an additional Drosophila AZ protein, "Blobby", essential for proper AZ nano-organization. Blobby biochemically associates with the ELKS family AZ scaffold protein Bruchpilot (BRP) and integrates into newly forming AZs. Loss of Blobby results in fewer AZs forming, ectopic AZ scaffold protein accumulations ("blobs") and disrupts nanoscale architecture of the BRP-AZ scaffold. Functionally, blobby mutants show diminished evoked synaptic currents due to reduced synaptic vesicle release probability and fewer functional release sites. Blobby is also present in adult brain synapses, and post-developmental knockdown of Blobby in the mushroom body impairs olfactory aversive memory consolidation. Thus, our analysis identifies an additional layer of AZ regulation critical for developmental AZ assembly but also for AZ-mediated plasticity controlling behavior.
Collapse
Affiliation(s)
- J Lützkendorf
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Matkovic-Rachid
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - T Götz
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - L Gao
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - O Turrel
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - M Maglione
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, SupraFAB, Berlin, Germany
| | - M Grieger
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Putignano
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Ramesh
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Ghelani
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A Neumann
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Gimber
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - J Schmoranzer
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - A Stawrakakis
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - B Brence
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - D Baum
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - Kai Ludwig
- Freie Universität Berlin, Institut für Chemie and Biochemie, Forschungszentrum für Elektronenmikroskopie, Berlin, Germany
| | - M Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Service Group, Berlin, Germany
| | - F Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
- University of Copenhagen, Department of Neuroscience, Copenhagen, Denmark
| | - M C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - S J Sigrist
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany.
- Charité Universitätsmedizin, NeuroCure Cluster of Excellence, Charitéplatz, Berlin, Germany.
| |
Collapse
|
5
|
Medeiros AT, Gratz SJ, Delgado A, Ritt JT, O'Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. eLife 2024; 12:RP88412. [PMID: 39291956 PMCID: PMC11410372 DOI: 10.7554/elife.88412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- Audrey T Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, United States
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, United States
| | - Ambar Delgado
- Department of Neuroscience, Brown University, Providence, United States
| | - Jason T Ritt
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| | - Kate M O'Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, United States
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| |
Collapse
|
6
|
Chien C, He K, Perry S, Tchitchkan E, Han Y, Li X, Dickman D. Distinct input-specific mechanisms enable presynaptic homeostatic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612361. [PMID: 39314403 PMCID: PMC11419068 DOI: 10.1101/2024.09.10.612361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Synapses are endowed with the flexibility to change through experience, but must be sufficiently stable to last a lifetime. This tension is illustrated at the Drosophila neuromuscular junction (NMJ), where two motor inputs that differ in structural and functional properties co-innervate most muscles to coordinate locomotion. To stabilize NMJ activity, motor neurons augment neurotransmitter release following diminished postsynaptic glutamate receptor functionality, termed presynaptic homeostatic potentiation (PHP). How these distinct inputs contribute to PHP plasticity remains enigmatic. We have used a botulinum neurotoxin to selectively silence each input and resolve their roles in PHP, demonstrating that PHP is input-specific: Chronic (genetic) PHP selectively targets the tonic MN-Ib, where active zone remodeling enhances Ca2+ influx to promote increased glutamate release. In contrast, acute (pharmacological) PHP selectively increases vesicle pools to potentiate phasic MN-Is. Thus, distinct homeostatic modulations in active zone nanoarchitecture, vesicle pools, and Ca2+ influx collaborate to enable input-specific PHP expression.
Collapse
Affiliation(s)
- Chun Chien
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Kaikai He
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Sarah Perry
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| | - Elizabeth Tchitchkan
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| | - Yifu Han
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Xiling Li
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Dion Dickman
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| |
Collapse
|
7
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Distinct active zone protein machineries mediate Ca 2+ channel clustering and vesicle priming at hippocampal synapses. Nat Neurosci 2024; 27:1680-1694. [PMID: 39160372 PMCID: PMC11682530 DOI: 10.1038/s41593-024-01720-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/28/2024] [Indexed: 08/21/2024]
Abstract
Action potentials trigger neurotransmitter release at the presynaptic active zone with spatiotemporal precision. This is supported by protein machinery that mediates synaptic vesicle priming and clustering of CaV2 Ca2+ channels nearby. One model posits that scaffolding proteins directly tether vesicles to CaV2s; however, here we find that at mouse hippocampal synapses, CaV2 clustering and vesicle priming are executed by separate machineries. CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins but distinct interaction motifs independently execute these functions. In transfected cells, Liprin-α and RIM form co-assemblies that are separate from CaV2-organizing complexes. At synapses, Liprin-α1-Liprin-α4 knockout impairs vesicle priming but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering CaV2s. We conclude that active zones consist of distinct machineries to organize CaV2s and prime vesicles, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Lindborg SR, Goyal NA, Katz J, Burford M, Li J, Kaspi H, Abramov N, Boulanger B, Berry JD, Nicholson K, Mozaffar T, Miller R, Jenkins L, Baloh RH, Lewis R, Staff NP, Owegi MA, Dagher B, Blondheim-Shraga NR, Gothelf Y, Levy YS, Kern R, Aricha R, Windebank AJ, Bowser R, Brown RH, Cudkowicz ME. Debamestrocel multimodal effects on biomarker pathways in amyotrophic lateral sclerosis are linked to clinical outcomes. Muscle Nerve 2024; 69:719-729. [PMID: 38593477 DOI: 10.1002/mus.28093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION/AIMS Biomarkers have shown promise in amyotrophic lateral sclerosis (ALS) research, but the quest for reliable biomarkers remains active. This study evaluates the effect of debamestrocel on cerebrospinal fluid (CSF) biomarkers, an exploratory endpoint. METHODS A total of 196 participants randomly received debamestrocel or placebo. Seven CSF samples were to be collected from all participants. Forty-five biomarkers were analyzed in the overall study and by two subgroups characterized by the ALS Functional Rating Scale-Revised (ALSFRS-R). A prespecified model was employed to predict clinical outcomes leveraging biomarkers and disease characteristics. Causal inference was used to analyze relationships between neurofilament light chain (NfL) and ALSFRS-R. RESULTS We observed significant changes with debamestrocel in 64% of the biomarkers studied, spanning pathways implicated in ALS pathology (63% neuroinflammation, 50% neurodegeneration, and 89% neuroprotection). Biomarker changes with debamestrocel show biological activity in trial participants, including those with advanced ALS. CSF biomarkers were predictive of clinical outcomes in debamestrocel-treated participants (baseline NfL, baseline latency-associated peptide/transforming growth factor beta1 [LAP/TGFβ1], change galectin-1, all p < .01), with baseline NfL and LAP/TGFβ1 remaining (p < .05) when disease characteristics (p < .005) were incorporated. Change from baseline to the last measurement showed debamestrocel-driven reductions in NfL were associated with less decline in ALSFRS-R. Debamestrocel significantly reduced NfL from baseline compared with placebo (11% vs. 1.6%, p = .037). DISCUSSION Following debamestrocel treatment, many biomarkers showed increases (anti-inflammatory/neuroprotective) or decreases (inflammatory/neurodegenerative) suggesting a possible treatment effect. Neuroinflammatory and neuroprotective biomarkers were predictive of clinical response, suggesting a potential multimodal mechanism of action. These results offer preliminary insights that need to be confirmed.
Collapse
Affiliation(s)
| | - Namita A Goyal
- UCI Health ALS & Neuromuscular Center, University of California, Irvine, California, USA
| | - Jonathan Katz
- Sutter Pacific Medical Foundation, California Pacific Medical Center, San Francisco, California, USA
| | - Matthew Burford
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jenny Li
- Brainstorm Cell Therapeutics, Boston, Massachusetts, USA
| | | | | | - Bruno Boulanger
- Department of Statistics and Data Science, PharmaLex, Mont-Saint-Guibert, Belgium
| | - James D Berry
- Healey & AMG Center, Mass General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Katharine Nicholson
- Healey & AMG Center, Mass General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tahseen Mozaffar
- UCI Health ALS & Neuromuscular Center, University of California, Irvine, California, USA
| | - Robert Miller
- Sutter Pacific Medical Foundation, California Pacific Medical Center, San Francisco, California, USA
| | - Liberty Jenkins
- Sutter Pacific Medical Foundation, California Pacific Medical Center, San Francisco, California, USA
| | - Robert H Baloh
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Richard Lewis
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nathan P Staff
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Margaret Ayo Owegi
- Neurology Department, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Bob Dagher
- Brainstorm Cell Therapeutics, Boston, Massachusetts, USA
| | | | | | - Yossef S Levy
- Manufacturing, Brainstorm Cell Therapeutics, Tel Aviv, Israel
| | - Ralph Kern
- Brainstorm Cell Therapeutics, Boston, Massachusetts, USA
| | | | - Anthony J Windebank
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Robert Bowser
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Robert H Brown
- Neurology Department, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Merit E Cudkowicz
- Healey & AMG Center, Mass General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Medeiros AT, Gratz S, Delgado A, Ritt J, O’Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535290. [PMID: 37034654 PMCID: PMC10081318 DOI: 10.1101/2023.04.02.535290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- A. T. Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, RI
| | - S.J. Gratz
- Department of Neuroscience, Brown University, Providence, RI
| | - A. Delgado
- Department of Neuroscience, Brown University, Providence, RI
| | - J.T. Ritt
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| | - Kate M. O’Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, RI
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| |
Collapse
|
10
|
Crane AB, Jetti SK, Littleton JT. A stochastic RNA editing process targets a limited number of sites in individual Drosophila glutamatergic motoneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594696. [PMID: 38798345 PMCID: PMC11118563 DOI: 10.1101/2024.05.17.594696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
RNA editing is a post-transcriptional source of protein diversity and occurs across the animal kingdom. Given the complete profile of mRNA targets and their editing rate in individual cells is unclear, we analyzed single cell RNA transcriptomes from Drosophila larval tonic and phasic glutamatergic motoneuron subtypes to determine the most highly edited targets and identify cell-type specific editing. From ∼15,000 genes encoded in the genome, 316 high confidence A-to-I canonical RNA edit sites were identified, with 102 causing missense amino acid changes in proteins regulating membrane excitability, synaptic transmission, and cellular function. Some sites showed 100% editing in single neurons as observed with mRNAs encoding mammalian AMPA receptors. However, most sites were edited at lower levels and generated variable expression of edited and unedited mRNAs within individual neurons. Together, these data provide insights into how the RNA editing landscape alters protein function to modulate the properties of two well-characterized neuronal populations in Drosophila .
Collapse
|
11
|
Turrel O, Gao L, Sigrist SJ. Presynaptic regulators in memory formation. Learn Mem 2024; 31:a054013. [PMID: 38862173 PMCID: PMC11199941 DOI: 10.1101/lm.054013.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 06/13/2024]
Abstract
The intricate molecular and structural sequences guiding the formation and consolidation of memories within neuronal circuits remain largely elusive. In this study, we investigate the roles of two pivotal presynaptic regulators, the small GTPase Rab3, enriched at synaptic vesicles, and the cell adhesion protein Neurexin-1, in the formation of distinct memory phases within the Drosophila mushroom body Kenyon cells. Our findings suggest that both proteins play crucial roles in memory-supporting processes within the presynaptic terminal, operating within distinct plasticity modules. These modules likely encompass remodeling and maturation of existing active zones (AZs), as well as the formation of new AZs.
Collapse
Affiliation(s)
- Oriane Turrel
- Institute for Biology, Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Lili Gao
- Institute for Biology, Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology, Genetics, Freie Universität Berlin, 14195 Berlin, Germany
- Cluster of Excellence NeuroCure, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
12
|
Agrawal B, Boulos S, Khatib S, Feuermann Y, Panov J, Kaphzan H. Molecular Insights into Transcranial Direct Current Stimulation Effects: Metabolomics and Transcriptomics Analyses. Cells 2024; 13:205. [PMID: 38334596 PMCID: PMC10854682 DOI: 10.3390/cells13030205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
INTRODUCTION Transcranial direct current stimulation (tDCS) is an evolving non-invasive neurostimulation technique. Despite multiple studies, its underlying molecular mechanisms are still unclear. Several previous human studies of the effect of tDCS suggest that it generates metabolic effects. The induction of metabolic effects by tDCS could provide an explanation for how it generates its long-term beneficial clinical outcome. AIM Given these hints of tDCS metabolic effects, we aimed to delineate the metabolic pathways involved in its mode of action. METHODS To accomplish this, we utilized a broad analytical approach of co-analyzing metabolomics and transcriptomic data generated from anodal tDCS in rat models. Since no metabolomic dataset was available, we performed a tDCS experiment of bilateral anodal stimulation of 200 µA for 20 min and for 5 consecutive days, followed by harvesting the brain tissue below the stimulating electrode and generating a metabolomics dataset using LC-MS/MS. The analysis of the transcriptomic dataset was based on a publicly available dataset. RESULTS Our analyses revealed that tDCS alters the metabolic profile of brain tissue, affecting bioenergetic-related pathways, such as glycolysis and mitochondrial functioning. In addition, we found changes in calcium-related signaling. CONCLUSIONS We conclude that tDCS affects metabolism by modulating energy production-related processes. Given our findings concerning calcium-related signaling, we suggest that the immediate effects of tDCS on calcium dynamics drive modifications in distinct metabolic pathways. A thorough understanding of the underlying molecular mechanisms of tDCS has the potential to revolutionize its applicability, enabling the generation of personalized medicine in the field of neurostimulation and thus contributing to its optimization.
Collapse
Affiliation(s)
- Bhanumita Agrawal
- Sagol Department of Neurobiology, University of Haifa, Haifa 3103301, Israel
| | - Soad Boulos
- Sagol Department of Neurobiology, University of Haifa, Haifa 3103301, Israel
| | - Soliman Khatib
- Department of Biotechnology, Tel-Hai College, Upper Galilee 1220800, Israel
| | - Yonatan Feuermann
- Sagol Department of Neurobiology, University of Haifa, Haifa 3103301, Israel
| | - Julia Panov
- Sagol Department of Neurobiology, University of Haifa, Haifa 3103301, Israel
- Tauber Bioinformatics Research Center, University of Haifa, Haifa 3103301, Israel
| | - Hanoch Kaphzan
- Sagol Department of Neurobiology, University of Haifa, Haifa 3103301, Israel
- Tauber Bioinformatics Research Center, University of Haifa, Haifa 3103301, Israel
| |
Collapse
|
13
|
Silva M, Tran V, Marty A. A maximum of two readily releasable vesicles per docking site at a cerebellar single active zone synapse. eLife 2024; 12:RP91087. [PMID: 38180320 PMCID: PMC10963025 DOI: 10.7554/elife.91087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024] Open
Abstract
Recent research suggests that in central mammalian synapses, active zones contain several docking sites acting in parallel. Before release, one or several synaptic vesicles (SVs) are thought to bind to each docking site, forming the readily releasable pool (RRP). Determining the RRP size per docking site has important implications for short-term synaptic plasticity. Here, using mouse cerebellar slices, we take advantage of recently developed methods to count the number of released SVs at single glutamatergic synapses in response to trains of action potentials (APs). In each recording, the number of docking sites was determined by fitting with a binomial model the number of released SVs in response to individual APs. After normalization with respect to the number of docking sites, the summed number of released SVs following a train of APs was used to estimate of the RRP size per docking site. To improve this estimate, various steps were taken to maximize the release probability of docked SVs, the occupancy of docking sites, as well as the extent of synaptic depression. Under these conditions, the RRP size reached a maximum value close to two SVs per docking site. The results indicate that each docking site contains two distinct SV-binding sites that can simultaneously accommodate up to one SV each. They further suggest that under special experimental conditions, as both sites are close to full occupancy, a maximal RRP size of two SVs per docking site can be reached. More generally, the results validate a sequential two-step docking model previously proposed at this preparation.
Collapse
Affiliation(s)
- Melissa Silva
- Université Paris Cité, SPPIN-Saints Pères Paris Institute for the Neurosciences, CNRSParisFrance
| | - Van Tran
- Université Paris Cité, SPPIN-Saints Pères Paris Institute for the Neurosciences, CNRSParisFrance
| | - Alain Marty
- Université Paris Cité, SPPIN-Saints Pères Paris Institute for the Neurosciences, CNRSParisFrance
| |
Collapse
|
14
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Molecular definition of distinct active zone protein machineries for Ca 2+ channel clustering and synaptic vesicle priming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564439. [PMID: 37961089 PMCID: PMC10634917 DOI: 10.1101/2023.10.27.564439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Action potentials trigger neurotransmitter release with minimal delay. Active zones mediate this temporal precision by co-organizing primed vesicles with CaV2 Ca2+ channels. The presumed model is that scaffolding proteins directly tether primed vesicles to CaV2s. We find that CaV2 clustering and vesicle priming are executed by separate machineries. At hippocampal synapses, CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins, but distinct interaction motifs independently execute these functions. In heterologous cells, Liprin-α and RIM from co-assemblies that are separate from CaV2-organizing complexes upon co-transfection. At synapses, Liprin-α1-4 knockout impairs vesicle priming, but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering of CaV2s. We conclude that active zones consist of distinct complexes to organize CaV2s and vesicle priming, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R. Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Aaron D. Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Poorna A. Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | |
Collapse
|
15
|
Willemse SW, Harley P, van Eijk RPA, Demaegd KC, Zelina P, Pasterkamp RJ, van Damme P, Ingre C, van Rheenen W, Veldink JH, Kiernan MC, Al-Chalabi A, van den Berg LH, Fratta P, van Es MA. UNC13A in amyotrophic lateral sclerosis: from genetic association to therapeutic target. J Neurol Neurosurg Psychiatry 2023; 94:649-656. [PMID: 36737245 PMCID: PMC10359588 DOI: 10.1136/jnnp-2022-330504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with limited treatment options and an incompletely understood pathophysiology. Although genomewide association studies (GWAS) have advanced our understanding of the disease, the precise manner in which risk polymorphisms contribute to disease pathogenesis remains unclear. Of relevance, GWAS have shown that a polymorphism (rs12608932) in the UNC13A gene is associated with risk for both ALS and frontotemporal dementia (FTD). Homozygosity for the C-allele at rs12608932 modifies the ALS phenotype, as these patients are more likely to have bulbar-onset disease, cognitive impairment and FTD at baseline as well as shorter survival. UNC13A is expressed in neuronal tissue and is involved in maintaining synaptic active zones, by enabling the priming and docking of synaptic vesicles. In the absence of functional TDP-43, risk variants in UNC13A lead to the inclusion of a cryptic exon in UNC13A messenger RNA, subsequently leading to nonsense mediated decay, with loss of functional protein. Depletion of UNC13A leads to impaired neurotransmission. Recent discoveries have identified UNC13A as a potential target for therapy development in ALS, with a confirmatory trial with lithium carbonate in UNC13A cases now underway and future approaches with antisense oligonucleotides currently under consideration. Considering UNC13A is a potent phenotypic modifier, it may also impact clinical trial outcomes. This present review describes the path from the initial discovery of UNC13A as a risk gene in ALS to the current therapeutic options being explored and how knowledge of its distinct phenotype needs to be taken into account in future trials.
Collapse
Affiliation(s)
- Sean W Willemse
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Peter Harley
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Ruben P A van Eijk
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Koen C Demaegd
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Philip van Damme
- Department of Neurology, KU Leuven Hospital, Leuven, Belgium
- Laboratory of Neurobiology, VIB KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Caroline Ingre
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Matthew C Kiernan
- Bushell Chair of Neurology, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Neurology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Pietro Fratta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Michael A van Es
- Department of Neurology, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, The Netherlands
| |
Collapse
|
16
|
Jusyte M, Blaum N, Böhme MA, Berns MMM, Bonard AE, Vámosi ÁB, Pushpalatha KV, Kobbersmed JRL, Walter AM. Unc13A dynamically stabilizes vesicle priming at synaptic release sites for short-term facilitation and homeostatic potentiation. Cell Rep 2023; 42:112541. [PMID: 37243591 DOI: 10.1016/j.celrep.2023.112541] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/10/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023] Open
Abstract
Presynaptic plasticity adjusts neurotransmitter (NT) liberation. Short-term facilitation (STF) tunes synapses to millisecond repetitive activation, while presynaptic homeostatic potentiation (PHP) of NT release stabilizes transmission over minutes. Despite different timescales of STF and PHP, our analysis of Drosophila neuromuscular junctions reveals functional overlap and shared molecular dependence on the release-site protein Unc13A. Mutating Unc13A's calmodulin binding domain (CaM-domain) increases baseline transmission while blocking STF and PHP. Mathematical modeling suggests that Ca2+/calmodulin/Unc13A interaction plastically stabilizes vesicle priming at release sites and that CaM-domain mutation causes constitutive stabilization, thereby blocking plasticity. Labeling the functionally essential Unc13A MUN domain reveals higher STED microscopy signals closer to release sites following CaM-domain mutation. Acute phorbol ester treatment similarly enhances NT release and blocks STF/PHP in synapses expressing wild-type Unc13A, while CaM-domain mutation occludes this, indicating common downstream effects. Thus, Unc13A regulatory domains integrate signals across timescales to switch release-site participation for synaptic plasticity.
Collapse
Affiliation(s)
- Meida Jusyte
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Natalie Blaum
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mathias A Böhme
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Manon M M Berns
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Alix E Bonard
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Ábel B Vámosi
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | - Janus R L Kobbersmed
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Alexander M Walter
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
17
|
He K, Han Y, Li X, Hernandez RX, Riboul DV, Feghhi T, Justs KA, Mahneva O, Perry S, Macleod GT, Dickman D. Physiologic and Nanoscale Distinctions Define Glutamatergic Synapses in Tonic vs Phasic Neurons. J Neurosci 2023; 43:4598-4611. [PMID: 37221096 PMCID: PMC10286941 DOI: 10.1523/jneurosci.0046-23.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Neurons exhibit a striking degree of functional diversity, each one tuned to the needs of the circuitry in which it is embedded. A fundamental functional dichotomy occurs in activity patterns, with some neurons firing at a relatively constant "tonic" rate, while others fire in bursts, a "phasic" pattern. Synapses formed by tonic versus phasic neurons are also functionally differentiated, yet the bases of their distinctive properties remain enigmatic. A major challenge toward illuminating the synaptic differences between tonic and phasic neurons is the difficulty in isolating their physiological properties. At the Drosophila neuromuscular junction, most muscle fibers are coinnervated by two motor neurons: the tonic "MN-Ib" and phasic "MN-Is." Here, we used selective expression of a newly developed botulinum neurotoxin transgene to silence tonic or phasic motor neurons in Drosophila larvae of either sex. This approach highlighted major differences in their neurotransmitter release properties, including probability, short-term plasticity, and vesicle pools. Furthermore, Ca2+ imaging demonstrated ∼2-fold greater Ca2+ influx at phasic neuron release sites relative to tonic, along with an enhanced synaptic vesicle coupling. Finally, confocal and super-resolution imaging revealed that phasic neuron release sites are organized in a more compact arrangement, with enhanced stoichiometry of voltage-gated Ca2+ channels relative to other active zone scaffolds. These data suggest that distinctions in active zone nano-architecture and Ca2+ influx collaborate to differentially tune glutamate release at tonic versus phasic synaptic subtypes.SIGNIFICANCE STATEMENT "Tonic" and "phasic" neuronal subtypes, based on differential firing properties, are common across many nervous systems. Using a recently developed approach to selectively silence transmission from one of these two neurons, we reveal specialized synaptic functional and structural properties that distinguish these specialized neurons. This study provides important insights into how input-specific synaptic diversity is achieved, which could have implications for neurologic disorders that involve changes in synaptic function.
Collapse
Affiliation(s)
- Kaikai He
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
- USC Neuroscience Graduate Program, Los Angeles, California 90089
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
- USC Neuroscience Graduate Program, Los Angeles, California 90089
| | - Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
- USC Neuroscience Graduate Program, Los Angeles, California 90089
| | - Roberto X Hernandez
- Integrative Biology and Neuroscience Graduate Program, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431
- International Max Planck Research School for Brain and Behavior, Jupiter, Florida 33458
| | - Danielle V Riboul
- Integrative Biology Graduate Program, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431
| | - Touhid Feghhi
- Department of Physics, Florida Atlantic University, Boca Raton, Florida 33431
| | - Karlis A Justs
- Integrative Biology and Neuroscience Graduate Program, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431
| | - Olena Mahneva
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida 33458
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
| | - Gregory T Macleod
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida 33458
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, Florida 33458
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
18
|
Boyd RJ, McClymont SA, Barrientos NB, Hook PW, Law WD, Rose RJ, Waite EL, Rathinavelu J, Avramopoulos D, McCallion AS. Evaluating the mouse neural precursor line, SN4741, as a suitable proxy for midbrain dopaminergic neurons. BMC Genomics 2023; 24:306. [PMID: 37286935 PMCID: PMC10245633 DOI: 10.1186/s12864-023-09398-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023] Open
Abstract
To overcome the ethical and technical limitations of in vivo human disease models, the broader scientific community frequently employs model organism-derived cell lines to investigate disease mechanisms, pathways, and therapeutic strategies. Despite the widespread use of certain in vitro models, many still lack contemporary genomic analysis supporting their use as a proxy for the affected human cells and tissues. Consequently, it is imperative to determine how accurately and effectively any proposed biological surrogate may reflect the biological processes it is assumed to model. One such cellular surrogate of human disease is the established mouse neural precursor cell line, SN4741, which has been used to elucidate mechanisms of neurotoxicity in Parkinson disease for over 25 years. Here, we are using a combination of classic and contemporary genomic techniques - karyotyping, RT-qPCR, single cell RNA-seq, bulk RNA-seq, and ATAC-seq - to characterize the transcriptional landscape, chromatin landscape, and genomic architecture of this cell line, and evaluate its suitability as a proxy for midbrain dopaminergic neurons in the study of Parkinson disease. We find that SN4741 cells possess an unstable triploidy and consistently exhibits low expression of dopaminergic neuron markers across assays, even when the cell line is shifted to the non-permissive temperature that drives differentiation. The transcriptional signatures of SN4741 cells suggest that they are maintained in an undifferentiated state at the permissive temperature and differentiate into immature neurons at the non-permissive temperature; however, they may not be dopaminergic neuron precursors, as previously suggested. Additionally, the chromatin landscapes of SN4741 cells, in both the differentiated and undifferentiated states, are not concordant with the open chromatin profiles of ex vivo, mouse E15.5 forebrain- or midbrain-derived dopaminergic neurons. Overall, our data suggest that SN4741 cells may reflect early aspects of neuronal differentiation but are likely not a suitable proxy for dopaminergic neurons as previously thought. The implications of this study extend broadly, illuminating the need for robust biological and genomic rationale underpinning the use of in vitro models of molecular processes.
Collapse
Affiliation(s)
- Rachel J. Boyd
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Sarah A. McClymont
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Nelson B. Barrientos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Paul W. Hook
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - William D. Law
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Rebecca J. Rose
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Eric L. Waite
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Jay Rathinavelu
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Dimitrios Avramopoulos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Andrew S. McCallion
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| |
Collapse
|
19
|
Krout M, Oh KH, Xiong A, Frankel EB, Kurshan PT, Kim H, Richmond JE. C. elegans Clarinet/CLA-1 recruits RIMB-1/RIM-binding protein and UNC-13 to orchestrate presynaptic neurotransmitter release. Proc Natl Acad Sci U S A 2023; 120:e2220856120. [PMID: 37186867 PMCID: PMC10214197 DOI: 10.1073/pnas.2220856120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Synaptic transmission requires the coordinated activity of multiple synaptic proteins that are localized at the active zone (AZ). We previously identified a Caenorhabditis elegans protein named Clarinet (CLA-1) based on homology to the AZ proteins Piccolo, Rab3-interactingmolecule (RIM)/UNC-10 and Fife. At the neuromuscular junction (NMJ), cla-1 null mutants exhibit release defects that are greatly exacerbated in cla-1;unc-10 double mutants. To gain insights into the coordinated roles of CLA-1 and UNC-10, we examined the relative contributions of each to the function and organization of the AZ. Using a combination of electrophysiology, electron microscopy, and quantitative fluorescence imaging we explored the functional relationship of CLA-1 to other key AZ proteins including: RIM1, Cav2.1 channels, RIM1-binding protein, and Munc13 (C. elegans UNC-10, UNC-2, RIMB-1 and UNC-13, respectively). Our analyses show that CLA-1 acts in concert with UNC-10 to regulate UNC-2 calcium channel levels at the synapse via recruitment of RIMB-1. In addition, CLA-1 exerts a RIMB-1-independent role in the localization of the priming factor UNC-13. Thus C. elegans CLA-1/UNC-10 exhibit combinatorial effects that have overlapping design principles with other model organisms: RIM/RBP and RIM/ELKS in mouse and Fife/RIM and BRP/RBP in Drosophila. These data support a semiconserved arrangement of AZ scaffolding proteins that are necessary for the localization and activation of the fusion machinery within nanodomains for precise coupling to Ca2+ channels.
Collapse
Affiliation(s)
- Mia Krout
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
| | - Kelly H. Oh
- Department of Cell Biology and Anatomy, Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Ame Xiong
- Department of Cell Biology and Anatomy, Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Elisa B. Frankel
- Department of Genetics, Albert Einstein College of Medicine, New York, NY10461
| | - Peri T. Kurshan
- Department of Genetics, Albert Einstein College of Medicine, New York, NY10461
| | - Hongkyun Kim
- Department of Genetics, Albert Einstein College of Medicine, New York, NY10461
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
| |
Collapse
|
20
|
Grasskamp AT, Jusyte M, McCarthy AW, Götz TWB, Ditlevsen S, Walter AM. Spontaneous neurotransmission at evocable synapses predicts their responsiveness to action potentials. Front Cell Neurosci 2023; 17:1129417. [PMID: 36970416 PMCID: PMC10030884 DOI: 10.3389/fncel.2023.1129417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/16/2023] [Indexed: 03/29/2023] Open
Abstract
Synaptic transmission relies on presynaptic neurotransmitter (NT) release from synaptic vesicles (SVs) and on NT detection by postsynaptic receptors. Transmission exists in two principal modes: action-potential (AP) evoked and AP-independent, "spontaneous" transmission. AP-evoked neurotransmission is considered the primary mode of inter-neuronal communication, whereas spontaneous transmission is required for neuronal development, homeostasis, and plasticity. While some synapses appear dedicated to spontaneous transmission only, all AP-responsive synapses also engage spontaneously, but whether this encodes functional information regarding their excitability is unknown. Here we report on functional interdependence of both transmission modes at individual synaptic contacts of Drosophila larval neuromuscular junctions (NMJs) which were identified by the presynaptic scaffolding protein Bruchpilot (BRP) and whose activities were quantified using the genetically encoded Ca2+ indicator GCaMP. Consistent with the role of BRP in organizing the AP-dependent release machinery (voltage-dependent Ca2+ channels and SV fusion machinery), most active BRP-positive synapses (>85%) responded to APs. At these synapses, the level of spontaneous activity was a predictor for their responsiveness to AP-stimulation. AP-stimulation resulted in cross-depletion of spontaneous activity and both transmission modes were affected by the non-specific Ca2+ channel blocker cadmium and engaged overlapping postsynaptic receptors. Thus, by using overlapping machinery, spontaneous transmission is a continuous, stimulus independent predictor for the AP-responsiveness of individual synapses.
Collapse
Affiliation(s)
| | - Meida Jusyte
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Einstein Center for Neurosciences, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | | | - Torsten W. B. Götz
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Susanne Ditlevsen
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander M. Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Einstein Center for Neurosciences, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
Ghelani T, Escher M, Thomas U, Esch K, Lützkendorf J, Depner H, Maglione M, Parutto P, Gratz S, Matkovic-Rachid T, Ryglewski S, Walter AM, Holcman D, O‘Connor Giles K, Heine M, Sigrist SJ. Interactive nanocluster compaction of the ELKS scaffold and Cacophony Ca 2+ channels drives sustained active zone potentiation. SCIENCE ADVANCES 2023; 9:eade7804. [PMID: 36800417 PMCID: PMC9937578 DOI: 10.1126/sciadv.ade7804] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/17/2023] [Indexed: 06/01/2023]
Abstract
At presynaptic active zones (AZs), conserved scaffold protein architectures control synaptic vesicle (SV) release by defining the nanoscale distribution and density of voltage-gated Ca2+ channels (VGCCs). While AZs can potentiate SV release in the minutes range, we lack an understanding of how AZ scaffold components and VGCCs engage into potentiation. We here establish dynamic, intravital single-molecule imaging of endogenously tagged proteins at Drosophila AZs undergoing presynaptic homeostatic potentiation. During potentiation, the numbers of α1 VGCC subunit Cacophony (Cac) increased per AZ, while their mobility decreased and nanoscale distribution compacted. These dynamic Cac changes depended on the interaction between Cac channel's intracellular carboxyl terminus and the membrane-close amino-terminal region of the ELKS-family protein Bruchpilot, whose distribution compacted drastically. The Cac-ELKS/Bruchpilot interaction was also needed for sustained AZ potentiation. Our single-molecule analysis illustrates how the AZ scaffold couples to VGCC nanoscale distribution and dynamics to establish a state of sustained potentiation.
Collapse
Affiliation(s)
- Tina Ghelani
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Marc Escher
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Ulrich Thomas
- Department of Cellular Neurobiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Klara Esch
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Janine Lützkendorf
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Harald Depner
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Marta Maglione
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
- Institute for Chemistry and Biochemistry, SupraFAB, Freie Universität Berlin, Altensteinstr. 23a, 14195 Berlin, Germany
| | - Pierre Parutto
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Scott Gratz
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Tanja Matkovic-Rachid
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Stefanie Ryglewski
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander M. Walter
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- Department of Neuroscience, University of Copenhagen, Copenhagen 2200, Denmark
| | - David Holcman
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Kate O‘Connor Giles
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Group Molecular Physiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Stephan J. Sigrist
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
22
|
Boyd RJ, McClymont SA, Barrientos NB, Hook PW, Law WD, Rose RJ, Waite EL, Rathinavelu J, Avramopoulos D, McCallion AS. Evaluating the mouse neural precursor line, SN4741, as a suitable proxy for midbrain dopaminergic neurons. RESEARCH SQUARE 2023:rs.3.rs-2520557. [PMID: 36824793 PMCID: PMC9949168 DOI: 10.21203/rs.3.rs-2520557/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
To overcome the ethical and technical limitations of in vivo human disease models, the broader scientific community frequently employs model organism-derived cell lines to investigate of disease mechanisms, pathways, and therapeutic strategies. Despite the widespread use of certain in vitro models, many still lack contemporary genomic analysis supporting their use as a proxy for the affected human cells and tissues. Consequently, it is imperative to determine how accurately and effectively any proposed biological surrogate may reflect the biological processes it is assumed to model. One such cellular surrogate of human disease is the established mouse neural precursor cell line, SN4741, which has been used to elucidate mechanisms of neurotoxicity in Parkinson disease for over 25 years. Here, we are using a combination of classic and contemporary genomic techniques - karyotyping, RT-qPCR, single cell RNA-seq, bulk RNA-seq, and ATAC-seq - to characterize the transcriptional landscape, chromatin landscape, and genomic architecture of this cell line, and evaluate its suitability as a proxy for midbrain dopaminergic neurons in the study of Parkinson disease. We find that SN4741 cells possess an unstable triploidy and consistently exhibits low expression of dopaminergic neuron markers across assays, even when the cell line is shifted to the non-permissive temperature that drives differentiation. The transcriptional signatures of SN4741 cells suggest that they are maintained in an undifferentiated state at the permissive temperature and differentiate into immature neurons at the non-permissive temperature; however, they may not be dopaminergic neuron precursors, as previously suggested. Additionally, the chromatin landscapes of SN4741 cells, in both the differentiated and undifferentiated states, are not concordant with the open chromatin profiles of ex vivo , mouse E15.5 forebrain- or midbrain-derived dopaminergic neurons. Overall, our data suggest that SN4741 cells may reflect early aspects of neuronal differentiation but are likely not a suitable a proxy for dopaminergic neurons as previously thought. The implications of this study extend broadly, illuminating the need for robust biological and genomic rationale underpinning the use of in vitro models of molecular processes.
Collapse
|
23
|
Boyd RJ, McClymont SA, Barrientos NB, Hook PW, Law WD, Rose RJ, Waite EL, Avramopoulos D, McCallion AS. Evaluating the mouse neural precursor line, SN4741, as a suitable proxy for midbrain dopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525270. [PMID: 36747739 PMCID: PMC9900784 DOI: 10.1101/2023.01.23.525270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
To overcome the ethical and technical limitations of in vivo human disease models, the broader scientific community frequently employs model organism-derived cell lines to investigate of disease mechanisms, pathways, and therapeutic strategies. Despite the widespread use of certain in vitro models, many still lack contemporary genomic analysis supporting their use as a proxy for the affected human cells and tissues. Consequently, it is imperative to determine how accurately and effectively any proposed biological surrogate may reflect the biological processes it is assumed to model. One such cellular surrogate of human disease is the established mouse neural precursor cell line, SN4741, which has been used to elucidate mechanisms of neurotoxicity in Parkinson disease for over 25 years. Here, we are using a combination of classic and contemporary genomic techniques - karyotyping, RT-qPCR, single cell RNA-seq, bulk RNA-seq, and ATAC-seq - to characterize the transcriptional landscape, chromatin landscape, and genomic architecture of this cell line, and evaluate its suitability as a proxy for midbrain dopaminergic neurons in the study of Parkinson disease. We find that SN4741 cells possess an unstable triploidy and consistently exhibits low expression of dopaminergic neuron markers across assays, even when the cell line is shifted to the non-permissive temperature that drives differentiation. The transcriptional signatures of SN4741 cells suggest that they are maintained in an undifferentiated state at the permissive temperature and differentiate into immature neurons at the non-permissive temperature; however, they may not be dopaminergic neuron precursors, as previously suggested. Additionally, the chromatin landscapes of SN4741 cells, in both the differentiated and undifferentiated states, are not concordant with the open chromatin profiles of ex vivo , mouse E15.5 forebrain- or midbrain-derived dopaminergic neurons. Overall, our data suggest that SN4741 cells may reflect early aspects of neuronal differentiation but are likely not a suitable a proxy for dopaminergic neurons as previously thought. The implications of this study extend broadly, illuminating the need for robust biological and genomic rationale underpinning the use of in vitro models of molecular processes.
Collapse
Affiliation(s)
- Rachel J. Boyd
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarah A. McClymont
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nelson B. Barrientos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul W. Hook
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - William D. Law
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rebecca J. Rose
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Eric L. Waite
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dimitrios Avramopoulos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew S. McCallion
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
24
|
Aldahabi M, Balint F, Holderith N, Lorincz A, Reva M, Nusser Z. Different priming states of synaptic vesicles underlie distinct release probabilities at hippocampal excitatory synapses. Neuron 2022; 110:4144-4161.e7. [PMID: 36261033 PMCID: PMC9796815 DOI: 10.1016/j.neuron.2022.09.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/28/2022] [Accepted: 09/27/2022] [Indexed: 12/12/2022]
Abstract
A stunning example of synaptic diversity is the postsynaptic target cell-type-dependent difference in synaptic efficacy in cortical networks. Here, we show that CA1 pyramidal cell (PC) to fast spiking interneuron (FSIN) connections have 10-fold larger release probability (Pv) than those on oriens lacunosum-moleculare (O-LM) interneurons. Freeze-fracture immunolabeling revealed that different nano-topologies and coupling distances between Ca2+ channels and release sites (RSs) are not responsible for the distinct Pv. Although [Ca2+] transients are 40% larger in FSINs innervating boutons, when [Ca2+] entry is matched in the two bouton populations, EPSCs in O-LM cells are still 7-fold smaller. However, application of a phorbol ester analog resulted in a ∼2.5-fold larger augmentation at PC - O-LM compared to PC - FSIN synapses, suggesting incomplete docking or priming of vesicles. Similar densities of docked vesicles rule out distinct RS occupancies and demonstrate that incompletely primed, but docked, vesicles limit the output of PC - O-LM synapses.
Collapse
Affiliation(s)
- Mohammad Aldahabi
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary,János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Flora Balint
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Noemi Holderith
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Andrea Lorincz
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Maria Reva
- Unit of Synapse and Circuit Dynamics, CNRS UMR 3571, Institute Pasteur, Paris, France
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary,Corresponding author
| |
Collapse
|
25
|
Dannhäuser S, Mrestani A, Gundelach F, Pauli M, Komma F, Kollmannsberger P, Sauer M, Heckmann M, Paul MM. Endogenous tagging of Unc-13 reveals nanoscale reorganization at active zones during presynaptic homeostatic potentiation. Front Cell Neurosci 2022; 16:1074304. [PMID: 36589286 PMCID: PMC9797049 DOI: 10.3389/fncel.2022.1074304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Neurotransmitter release at presynaptic active zones (AZs) requires concerted protein interactions within a dense 3D nano-hemisphere. Among the complex protein meshwork the (M)unc-13 family member Unc-13 of Drosophila melanogaster is essential for docking of synaptic vesicles and transmitter release. Methods We employ minos-mediated integration cassette (MiMIC)-based gene editing using GFSTF (EGFP-FlAsH-StrepII-TEV-3xFlag) to endogenously tag all annotated Drosophila Unc-13 isoforms enabling visualization of endogenous Unc-13 expression within the central and peripheral nervous system. Results and discussion Electrophysiological characterization using two-electrode voltage clamp (TEVC) reveals that evoked and spontaneous synaptic transmission remain unaffected in unc-13 GFSTF 3rd instar larvae and acute presynaptic homeostatic potentiation (PHP) can be induced at control levels. Furthermore, multi-color structured-illumination shows precise co-localization of Unc-13GFSTF, Bruchpilot, and GluRIIA-receptor subunits within the synaptic mesoscale. Localization microscopy in combination with HDBSCAN algorithms detect Unc-13GFSTF subclusters that move toward the AZ center during PHP with unaltered Unc-13GFSTF protein levels.
Collapse
Affiliation(s)
- Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Achmed Mrestani
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Neurology, Leipzig University Medical Center, Leipzig, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Florian Gundelach
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Center of Mental Health, Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Würzburg, Würzburg, Germany
| | - Martin Pauli
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Fabian Komma
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Mila M Paul
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Turrel O, Ramesh N, Escher MJF, Pooryasin A, Sigrist SJ. Transient active zone remodeling in the Drosophila mushroom body supports memory. Curr Biol 2022; 32:4900-4913.e4. [PMID: 36327980 DOI: 10.1016/j.cub.2022.10.017] [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: 12/16/2021] [Revised: 08/15/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
Abstract
Elucidating how the distinct components of synaptic plasticity dynamically orchestrate the distinct stages of memory acquisition and maintenance within neuronal networks remains a major challenge. Specifically, plasticity processes tuning the functional and also structural state of presynaptic active zone (AZ) release sites are widely observed in vertebrates and invertebrates, but their behavioral relevance remains mostly unclear. We here provide evidence that a transient upregulation of presynaptic AZ release site proteins supports aversive olfactory mid-term memory in the Drosophila mushroom body (MB). Upon paired aversive olfactory conditioning, AZ protein levels (ELKS-family BRP/(m)unc13-family release factor Unc13A) increased for a few hours with MB-lobe-specific dynamics. Kenyon cell (KC, intrinsic MB neurons)-specific knockdown (KD) of BRP did not affect aversive olfactory short-term memory (STM) but strongly suppressed aversive mid-term memory (MTM). Different proteins crucial for the transport of AZ biosynthetic precursors (transport adaptor Aplip1/Jip-1; kinesin motor IMAC/Unc104; small GTPase Arl8) were also specifically required for the formation of aversive olfactory MTM. Consistent with the merely transitory increase of AZ proteins, BRP KD did not interfere with the formation of aversive olfactory long-term memory (LTM; i.e., 1 day). Our data suggest that the remodeling of presynaptic AZ refines the MB circuitry after paired aversive conditioning, over a time window of a few hours, to display aversive olfactory memories.
Collapse
Affiliation(s)
- Oriane Turrel
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Niraja Ramesh
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Marc J F Escher
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Atefeh Pooryasin
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
27
|
Tan C, de Nola G, Qiao C, Imig C, Born RT, Brose N, Kaeser PS. Munc13 supports fusogenicity of non-docked vesicles at synapses with disrupted active zones. eLife 2022; 11:79077. [PMID: 36398873 PMCID: PMC9822248 DOI: 10.7554/elife.79077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/17/2022] [Indexed: 11/19/2022] Open
Abstract
Active zones consist of protein scaffolds that are tightly attached to the presynaptic plasma membrane. They dock and prime synaptic vesicles, couple them to voltage-gated Ca2+ channels, and direct neurotransmitter release toward postsynaptic receptor domains. Simultaneous RIM + ELKS ablation disrupts these scaffolds, abolishes vesicle docking, and removes active zone-targeted Munc13, but some vesicles remain releasable. To assess whether this enduring vesicular fusogenicity is mediated by non-active zone-anchored Munc13 or is Munc13-independent, we ablated Munc13-1 and Munc13-2 in addition to RIM + ELKS in mouse hippocampal neurons. The hextuple knockout synapses lacked docked vesicles, but other ultrastructural features were near-normal despite the strong genetic manipulation. Removing Munc13 in addition to RIM + ELKS impaired action potential-evoked vesicle fusion more strongly than RIM + ELKS knockout by further decreasing the releasable vesicle pool. Hence, Munc13 can support some fusogenicity without RIM and ELKS, and presynaptic recruitment of Munc13, even without active zone anchoring, suffices to generate some fusion-competent vesicles.
Collapse
Affiliation(s)
- Chao Tan
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Claire Qiao
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Cordelia Imig
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Richard T Born
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
28
|
Midorikawa M. Developmental and activity-dependent modulation of coupling distance between release site and Ca2+ channel. Front Cell Neurosci 2022; 16:1037721. [DOI: 10.3389/fncel.2022.1037721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Synapses are junctions between a presynaptic neuron and a postsynaptic cell specialized for fast and precise information transfer. The presynaptic terminal secretes neurotransmitters via exocytosis of synaptic vesicles. Exocytosis is a tightly regulated reaction that occurs within a millisecond of the arrival of an action potential. One crucial parameter in determining the characteristics of the transmitter release kinetics is the coupling distance between the release site and the Ca2+ channel. Still, the technical limitations have hindered detailed analysis from addressing how the coupling distance is regulated depending on the development or activity of the synapse. However, recent technical advances in electrophysiology and imaging are unveiling their different configurations in different conditions. Here, I will summarize developmental- and activity-dependent changes in the coupling distances revealed by recent studies.
Collapse
|
29
|
Rizo J, David G, Fealey ME, Jaczynska K. On the difficulties of characterizing weak protein interactions that are critical for neurotransmitter release. FEBS Open Bio 2022; 12:1912-1938. [PMID: 35986639 PMCID: PMC9623538 DOI: 10.1002/2211-5463.13473] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
The mechanism of neurotransmitter release has been extensively characterized, showing that vesicle fusion is mediated by the SNARE complex formed by syntaxin-1, SNAP-25 and synaptobrevin. This complex is disassembled by N-ethylmaleimide sensitive factor (NSF) and SNAPs to recycle the SNAREs, whereas Munc18-1 and Munc13s organize SNARE complex assembly in an NSF-SNAP-resistant manner. Synaptotagmin-1 acts as the Ca2+ sensor that triggers exocytosis in a tight interplay with the SNAREs and complexins. Here, we review technical aspects associated with investigation of protein interactions underlying these steps, which is hindered because the release machinery is assembled between two membranes and is highly dynamic. Moreover, weak interactions, which are difficult to characterize, play key roles in neurotransmitter release, for instance by lowering energy barriers that need to be overcome in this highly regulated process. We illustrate the crucial role that structural biology has played in uncovering mechanisms underlying neurotransmitter release, but also discuss the importance of considering the limitations of the techniques used, including lessons learned from research in our lab and others. In particular, we emphasize: (a) the promiscuity of some protein sequences, including membrane-binding regions that can mediate irrelevant interactions with proteins in the absence of their native targets; (b) the need to ensure that weak interactions observed in crystal structures are biologically relevant; and (c) the limitations of isothermal titration calorimetry to analyze weak interactions. Finally, we stress that even studies that required re-interpretation often helped to move the field forward by improving our understanding of the system and providing testable hypotheses.
Collapse
Affiliation(s)
- Josep Rizo
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Guillaume David
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Michael E. Fealey
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Klaudia Jaczynska
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
30
|
A sequential two-step priming scheme reproduces diversity in synaptic strength and short-term plasticity. Proc Natl Acad Sci U S A 2022; 119:e2207987119. [PMID: 35969787 PMCID: PMC9407230 DOI: 10.1073/pnas.2207987119] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Central nervous system synapses are diverse in strength and plasticity. Short-term plasticity has traditionally been evaluated with models postulating a single pool of functionally homogeneous fusion-competent synaptic vesicles. Many observations are not easily explainable by such simple models. We established and experimentally validated a scheme of synaptic vesicle priming consisting of two sequential and reversible steps of release–machinery assembly. This sequential two-step priming scheme faithfully reproduced plasticity at a glutamatergic model synapse. The proposed priming and fusion scheme was consistent with the measured mean responses and with the experimentally observed heterogeneity between synapses. Vesicle fusion probability was found to be relatively uniform among synapses, while the priming equilibrium at rest of mature versus immature vesicle priming states differed greatly. Glutamatergic synapses display variable strength and diverse short-term plasticity (STP), even for a given type of connection. Using nonnegative tensor factorization and conventional state modeling, we demonstrate that a kinetic scheme consisting of two sequential and reversible steps of release–machinery assembly and a final step of synaptic vesicle (SV) fusion reproduces STP and its diversity among synapses. Analyzing transmission at the calyx of Held synapses reveals that differences in synaptic strength and STP are not primarily caused by variable fusion probability (pfusion) but are determined by the fraction of docked synaptic vesicles equipped with a mature release machinery. Our simulations show that traditional quantal analysis methods do not necessarily report pfusion of SVs with a mature release machinery but reflect both pfusion and the distribution between mature and immature priming states at rest. Thus, the approach holds promise for a better mechanistic dissection of the roles of presynaptic proteins in the sequence of SV docking, two-step priming, and fusion. It suggests a mechanism for activity-induced redistribution of synaptic efficacy.
Collapse
|
31
|
Duhart JC, Mosca TJ. Genetic regulation of central synapse formation and organization in Drosophila melanogaster. Genetics 2022; 221:6597078. [PMID: 35652253 DOI: 10.1093/genetics/iyac078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/29/2022] [Indexed: 01/04/2023] Open
Abstract
A goal of modern neuroscience involves understanding how connections in the brain form and function. Such a knowledge is essential to inform how defects in the exquisite complexity of nervous system growth influence neurological disease. Studies of the nervous system in the fruit fly Drosophila melanogaster enabled the discovery of a wealth of molecular and genetic mechanisms underlying development of synapses-the specialized cell-to-cell connections that comprise the essential substrate for information flow and processing in the nervous system. For years, the major driver of knowledge was the neuromuscular junction due to its ease of examination. Analogous studies in the central nervous system lagged due to a lack of genetic accessibility of specific neuron classes, synaptic labels compatible with cell-type-specific access, and high resolution, quantitative imaging strategies. However, understanding how central synapses form remains a prerequisite to understanding brain development. In the last decade, a host of new tools and techniques extended genetic studies of synapse organization into central circuits to enhance our understanding of synapse formation, organization, and maturation. In this review, we consider the current state-of-the-field. We first discuss the tools, technologies, and strategies developed to visualize and quantify synapses in vivo in genetically identifiable neurons of the Drosophila central nervous system. Second, we explore how these tools enabled a clearer understanding of synaptic development and organization in the fly brain and the underlying molecular mechanisms of synapse formation. These studies establish the fly as a powerful in vivo genetic model that offers novel insights into neural development.
Collapse
Affiliation(s)
- Juan Carlos Duhart
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Timothy J Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
32
|
Re-examination of the determinants of synaptic strength from the perspective of superresolution imaging. Curr Opin Neurobiol 2022; 74:102540. [DOI: 10.1016/j.conb.2022.102540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/04/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022]
|
33
|
Abstract
Major recent advances and previous data have led to a plausible model of how key proteins mediate neurotransmitter release. In this model, the soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein (SNAP) receptor (SNARE) proteins syntaxin-1, SNAP-25, and synaptobrevin form tight complexes that bring the membranes together and are crucial for membrane fusion. NSF and SNAPs disassemble SNARE complexes and ensure that fusion occurs through an exquisitely regulated pathway that starts with Munc18-1 bound to a closed conformation of syntaxin-1. Munc18-1 also binds to synaptobrevin, forming a template to assemble the SNARE complex when Munc13-1 opens syntaxin-1 while bridging the vesicle and plasma membranes. Synaptotagmin-1 and complexin bind to partially assembled SNARE complexes, likely stabilizing them and preventing fusion until Ca2+ binding to synaptotagmin-1 causes dissociation from the SNARE complex and induces interactions with phospholipids that help trigger release. Although fundamental questions remain about the mechanism of membrane fusion, these advances provide a framework to investigate the mechanisms underlying presynaptic plasticity.
Collapse
Affiliation(s)
- Josep Rizo
- Departments of Biophysics, Biochemistry, and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA;
| |
Collapse
|
34
|
Tan C, Wang SSH, de Nola G, Kaeser PS. Rebuilding essential active zone functions within a synapse. Neuron 2022; 110:1498-1515.e8. [PMID: 35176221 PMCID: PMC9081183 DOI: 10.1016/j.neuron.2022.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 01/15/2023]
Abstract
Presynaptic active zones are molecular machines that control neurotransmitter secretion. They form sites for vesicle docking and priming and couple vesicles to Ca2+ entry for release triggering. The complexity of active zone machinery has made it challenging to determine its mechanisms in release. Simultaneous knockout of the active zone proteins RIM and ELKS disrupts active zone assembly, abolishes vesicle docking, and impairs release. We here rebuild docking, priming, and Ca2+ secretion coupling in these mutants without reinstating active zone networks. Re-expression of RIM zinc fingers recruited Munc13 to undocked vesicles and rendered the vesicles release competent. Action potential triggering of release was reconstituted by docking these primed vesicles to Ca2+ channels through attaching RIM zinc fingers to CaVβ4-subunits. Our work identifies an 80-kDa β4-Zn protein that bypasses the need for megadalton-sized secretory machines, establishes that fusion competence and docking are mechanistically separable, and defines RIM zinc finger-Munc13 complexes as hubs for active zone function.
Collapse
Affiliation(s)
- Chao Tan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Knodel MM, Dutta Roy R, Wittum G. Influence of T-Bar on Calcium Concentration Impacting Release Probability. Front Comput Neurosci 2022; 16:855746. [PMID: 35586479 PMCID: PMC9108211 DOI: 10.3389/fncom.2022.855746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/09/2022] [Indexed: 11/25/2022] Open
Abstract
The relation of form and function, namely the impact of the synaptic anatomy on calcium dynamics in the presynaptic bouton, is a major challenge of present (computational) neuroscience at a cellular level. The Drosophila larval neuromuscular junction (NMJ) is a simple model system, which allows studying basic effects in a rather simple way. This synapse harbors several special structures. In particular, in opposite to standard vertebrate synapses, the presynaptic boutons are rather large, and they have several presynaptic zones. In these zones, different types of anatomical structures are present. Some of the zones bear a so-called T-bar, a particular anatomical structure. The geometric form of the T-bar resembles the shape of the letter “T” or a table with one leg. When an action potential arises, calcium influx is triggered. The probability of vesicle docking and neurotransmitter release is superlinearly proportional to the concentration of calcium close to the vesicular release site. It is tempting to assume that the T-bar causes some sort of calcium accumulation and hence triggers a higher release probability and thus enhances neurotransmitter exocytosis. In order to study this influence in a quantitative manner, we constructed a typical T-bar geometry and compared the calcium concentration close to the active zones (AZs). We compared the case of synapses with and without T-bars. Indeed, we found a substantial influence of the T-bar structure on the presynaptic calcium concentrations close to the AZs, indicating that this anatomical structure increases vesicle release probability. Therefore, our study reveals how the T-bar zone implies a strong relation between form and function. Our study answers the question of experimental studies (namely “Wichmann and Sigrist, Journal of neurogenetics 2010”) concerning the sense of the anatomical structure of the T-bar.
Collapse
Affiliation(s)
- Markus M. Knodel
- Goethe Center for Scientific Computing (GCSC), Goethe Universität Frankfurt, Frankfurt, Germany
- *Correspondence: Markus M. Knodel ; orcid.org/0000-0001-8739-0803
| | | | - Gabriel Wittum
- Goethe Center for Scientific Computing (GCSC), Goethe Universität Frankfurt, Frankfurt, Germany
- Applied Mathematics and Computational Science, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| |
Collapse
|
36
|
Qin N, Chen Z, Xue R. A two-subpopulation model that reflects heterogeneity of large dense core vesicles in exocytosis. Cell Cycle 2022; 21:531-546. [PMID: 35067177 PMCID: PMC8942488 DOI: 10.1080/15384101.2022.2026576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Exocytosis of large dense core vesicles is responsible for hormone secretion in neuroendocrine cells. The population of primed vesicles ready to release upon cell excitation demonstrates large heterogeneity. However, there are currently no models that clearly reflect such heterogeneity. Here, we develop a novel model based on single vesicle release events from amperometry recordings of PC12 cells using carbon fiber microelectrode. In this model, releasable vesicles can be grouped into two subpopulations, namely, SP1 and SP2. SP1 vesicles replenish quickly, with kinetics of ~0.0368 s-1, but likely undergo slow fusion pore expansion (amperometric signals rise at ~2.5 pA/ms), while SP2 vesicles demonstrate slow replenishment (kinetics of ~0.0048 s-1) but prefer fast dilation of fusion pore, with an amperometric signal rising rate of ~9.1 pA/ms. Phorbol ester enlarges the size of SP2 partially via activation of protein kinase C and conveys SP1 vesicles into SP2. Inhibition of Rho GTPase-dependent actin rearrangement almost completely depletes SP2. We also propose that the phorbol ester-sensitive vesicle subpopulation (SP2) is analogous to the subset of superprimed synaptic vesicles in neurons. This model provides a meticulous description of the architecture of the readily releasable vesicle pool and elucidates the heterogeneity of the vesicle priming mechanism.
Collapse
Affiliation(s)
- Nan Qin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhixi Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Renhao Xue
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China,CONTACT Renhao Xue Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
37
|
Munc13 structural transitions and oligomers that may choreograph successive stages in vesicle priming for neurotransmitter release. Proc Natl Acad Sci U S A 2022; 119:2121259119. [PMID: 35135883 PMCID: PMC8851502 DOI: 10.1073/pnas.2121259119] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
The speed of neural information processing in the human central nervous system is ultimately determined by the speed of chemical transmission at synapses, because action potentials have relatively short distances to traverse. The release of synaptic vesicles containing neurotransmitters must therefore be remarkably fast as compared to other forms of membrane fusion. Six separate SNARE complexes cooperate to achieve this. But how can exactly six copies be assembled under every vesicle? Here we report that six copies of the key molecular chaperone that assembles the SNAREs can arrange themselves into a closed hexagon, providing the likely answer. How can exactly six SNARE complexes be assembled under each synaptic vesicle? Here we report cryo-EM crystal structures of the core domain of Munc13, the key chaperone that initiates SNAREpin assembly. The functional core of Munc13, consisting of C1–C2B–MUN–C2C (Munc13C) spontaneously crystallizes between phosphatidylserine-rich bilayers in two distinct conformations, each in a radically different oligomeric state. In the open conformation (state 1), Munc13C forms upright trimers that link the two bilayers, separating them by ∼21 nm. In the closed conformation, six copies of Munc13C interact to form a lateral hexamer elevated ∼14 nm above the bilayer. Open and closed conformations differ only by a rigid body rotation around a flexible hinge, which when performed cooperatively assembles Munc13 into a lateral hexamer (state 2) in which the key SNARE assembly-activating site of Munc13 is autoinhibited by its neighbor. We propose that each Munc13 in the lateral hexamer ultimately assembles a single SNAREpin, explaining how only and exactly six SNARE complexes are templated. We suggest that state 1 and state 2 may represent two successive states in the synaptic vesicle supply chain leading to “primed” ready-release vesicles in which SNAREpins are clamped and ready to release (state 3).
Collapse
|
38
|
Newman ZL, Bakshinskaya D, Schultz R, Kenny SJ, Moon S, Aghi K, Stanley C, Marnani N, Li R, Bleier J, Xu K, Isacoff EY. Determinants of synapse diversity revealed by super-resolution quantal transmission and active zone imaging. Nat Commun 2022; 13:229. [PMID: 35017509 PMCID: PMC8752601 DOI: 10.1038/s41467-021-27815-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 12/06/2021] [Indexed: 01/23/2023] Open
Abstract
Neural circuit function depends on the pattern of synaptic connections between neurons and the strength of those connections. Synaptic strength is determined by both postsynaptic sensitivity to neurotransmitter and the presynaptic probability of action potential evoked transmitter release (Pr). Whereas morphology and neurotransmitter receptor number indicate postsynaptic sensitivity, presynaptic indicators and the mechanism that sets Pr remain to be defined. To address this, we developed QuaSOR, a super-resolution method for determining Pr from quantal synaptic transmission imaging at hundreds of glutamatergic synapses at a time. We mapped the Pr onto super-resolution 3D molecular reconstructions of the presynaptic active zones (AZs) of the same synapses at the Drosophila larval neuromuscular junction (NMJ). We find that Pr varies greatly between synapses made by a single axon, quantify the contribution of key AZ proteins to Pr diversity and find that one of these, Complexin, suppresses spontaneous and evoked transmission differentially, thereby generating a spatial and quantitative mismatch between release modes. Transmission is thus regulated by the balance and nanoscale distribution of release-enhancing and suppressing presynaptic proteins to generate high signal-to-noise evoked transmission.
Collapse
Affiliation(s)
- Zachary L Newman
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Dariya Bakshinskaya
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Ryan Schultz
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Samuel J Kenny
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - Seonah Moon
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - Krisha Aghi
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Cherise Stanley
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Nadia Marnani
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Rachel Li
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Julia Bleier
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Ke Xu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA
- Molecular Biophysics and Integrated BioImaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
- Molecular Biophysics and Integrated BioImaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
- Weill Neurohub, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
39
|
Piao C, Sigrist SJ. (M)Unc13s in Active Zone Diversity: A Drosophila Perspective. Front Synaptic Neurosci 2022; 13:798204. [PMID: 35046788 PMCID: PMC8762327 DOI: 10.3389/fnsyn.2021.798204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 12/03/2022] Open
Abstract
The so-called active zones at pre-synaptic terminals are the ultimate filtering devices, which couple between action potential frequency and shape, and the information transferred to the post-synaptic neurons, finally tuning behaviors. Within active zones, the release of the synaptic vesicle operates from specialized “release sites.” The (M)Unc13 class of proteins is meant to define release sites topologically and biochemically, and diversity between Unc13-type release factor isoforms is suspected to steer diversity at active zones. The two major Unc13-type isoforms, namely, Unc13A and Unc13B, have recently been described from the molecular to the behavioral level, exploiting Drosophila being uniquely suited to causally link between these levels. The exact nanoscale distribution of voltage-gated Ca2+ channels relative to release sites (“coupling”) at pre-synaptic active zones fundamentally steers the release of the synaptic vesicle. Unc13A and B were found to be either tightly or loosely coupled across Drosophila synapses. In this review, we reported recent findings on diverse aspects of Drosophila Unc13A and B, importantly, their nano-topological distribution at active zones and their roles in release site generation, active zone assembly, and pre-synaptic homeostatic plasticity. We compared their stoichiometric composition at different synapse types, reviewing the correlation between nanoscale distribution of these two isoforms and release physiology and, finally, discuss how isoform-specific release components might drive the functional heterogeneity of synapses and encode discrete behavior.
Collapse
Affiliation(s)
- Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Stephan J. Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
- *Correspondence: Stephan J. Sigrist
| |
Collapse
|
40
|
Abstract
Fluorescence imaging techniques play a pivotal role in our understanding of the nervous system. The emergence of various super-resolution microscopy methods and specialized fluorescent probes enables direct insight into neuronal structure and protein arrangements in cellular subcompartments with so far unmatched resolution. Super-resolving visualization techniques in neurons unveil a novel understanding of cytoskeletal composition, distribution, motility, and signaling of membrane proteins, subsynaptic structure and function, and neuron-glia interaction. Well-defined molecular targets in autoimmune and neurodegenerative disease models provide excellent starting points for in-depth investigation of disease pathophysiology using novel and innovative imaging methodology. Application of super-resolution microscopy in human brain samples and for testing clinical biomarkers is still in its infancy but opens new opportunities for translational research in neurology and neuroscience. In this review, we describe how super-resolving microscopy has improved our understanding of neuronal and brain function and dysfunction in the last two decades.
Collapse
Affiliation(s)
- Christian Werner
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
41
|
Sauvola CW, Littleton JT. SNARE Regulatory Proteins in Synaptic Vesicle Fusion and Recycling. Front Mol Neurosci 2021; 14:733138. [PMID: 34421538 PMCID: PMC8377282 DOI: 10.3389/fnmol.2021.733138] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Membrane fusion is a universal feature of eukaryotic protein trafficking and is mediated by the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) family. SNARE proteins embedded in opposing membranes spontaneously assemble to drive membrane fusion and cargo exchange in vitro. Evolution has generated a diverse complement of SNARE regulatory proteins (SRPs) that ensure membrane fusion occurs at the right time and place in vivo. While a core set of SNAREs and SRPs are common to all eukaryotic cells, a specialized set of SRPs within neurons confer additional regulation to synaptic vesicle (SV) fusion. Neuronal communication is characterized by precise spatial and temporal control of SNARE dynamics within presynaptic subdomains specialized for neurotransmitter release. Action potential-elicited Ca2+ influx at these release sites triggers zippering of SNAREs embedded in the SV and plasma membrane to drive bilayer fusion and release of neurotransmitters that activate downstream targets. Here we discuss current models for how SRPs regulate SNARE dynamics and presynaptic output, emphasizing invertebrate genetic findings that advanced our understanding of SRP regulation of SV cycling.
Collapse
Affiliation(s)
- Chad W Sauvola
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
42
|
Huang S, Sigrist SJ. Presynaptic and postsynaptic long-term plasticity in sleep homeostasis. Curr Opin Neurobiol 2021; 69:1-10. [DOI: 10.1016/j.conb.2020.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 12/25/2022]
|
43
|
Szikora S, Görög P, Kozma C, Mihály J. Drosophila Models Rediscovered with Super-Resolution Microscopy. Cells 2021; 10:1924. [PMID: 34440693 PMCID: PMC8391832 DOI: 10.3390/cells10081924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 11/25/2022] Open
Abstract
With the advent of super-resolution microscopy, we gained a powerful toolbox to bridge the gap between the cellular- and molecular-level analysis of living organisms. Although nanoscopy is broadly applicable, classical model organisms, such as fruit flies, worms and mice, remained the leading subjects because combining the strength of sophisticated genetics, biochemistry and electrophysiology with the unparalleled resolution provided by super-resolution imaging appears as one of the most efficient approaches to understanding the basic cell biological questions and the molecular complexity of life. Here, we summarize the major nanoscopic techniques and illustrate how these approaches were used in Drosophila model systems to revisit a series of well-known cell biological phenomena. These investigations clearly demonstrate that instead of simply achieving an improvement in image quality, nanoscopy goes far beyond with its immense potential to discover novel structural and mechanistic aspects. With the examples of synaptic active zones, centrosomes and sarcomeres, we will explain the instrumental role of super-resolution imaging pioneered in Drosophila in understanding fundamental subcellular constituents.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
| | - Péter Görög
- Institute of Genetics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
- Doctoral School of Multidisciplinary Medical Science, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - Csaba Kozma
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, Pálfy u. 52/d, H-6725 Szeged, Hungary;
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary
| |
Collapse
|
44
|
Tracking Single Molecule Dynamics in the Adult Drosophila Brain. eNeuro 2021; 8:ENEURO.0057-21.2021. [PMID: 33875453 PMCID: PMC8174007 DOI: 10.1523/eneuro.0057-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 11/21/2022] Open
Abstract
Super-resolution microscopy provides valuable insight for understanding the nanoscale organization within living tissue, although this method is typically restricted to cultured or dissociated cells. Here, we develop a method to track the mobility of individual proteins in ex vivo adult Drosophila melanogaster brains, focusing on a key component of the presynaptic release machinery, syntaxin1A (Sx1a). We show that individual Sx1a dynamics can be reliably tracked within neurons in the whole fly brain, and that the mobility of Sx1a molecules increases following conditional neural stimulation. We then apply this preparation to the problem of general anesthesia, to address how different anesthetics might affect single molecule dynamics in intact brain synapses. We find that propofol, etomidate, and isoflurane significantly impair Sx1a mobility, while ketamine and sevoflurane have little effect. Resolving single molecule dynamics in intact fly brains provides a novel approach to link localized molecular effects with systems-level phenomena such as general anesthesia.
Collapse
|
45
|
Silva M, Tran V, Marty A. Calcium-dependent docking of synaptic vesicles. Trends Neurosci 2021; 44:579-592. [PMID: 34049722 DOI: 10.1016/j.tins.2021.04.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/23/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022]
Abstract
The concentration of calcium ions in presynaptic terminals regulates transmitter release, but underlying mechanisms have remained unclear. Here we review recent studies that shed new light on this issue. Fast-freezing electron microscopy and total internal reflection fluorescence microscopy studies reveal complex calcium-dependent vesicle movements including docking on a millisecond time scale. Recordings from so-called 'simple synapses' indicate that calcium not only triggers exocytosis, but also modifies synaptic strength by controlling a final, rapid vesicle maturation step before release. Molecular studies identify several calcium-sensitive domains on Munc13 and on synaptotagmin-1 that are likely involved in bringing the vesicular and plasma membranes closer together in response to calcium elevation. Together, these results suggest that calcium-dependent vesicle docking occurs in a wide range of time domains and plays a crucial role in several phenomena including synaptic facilitation, post-tetanic potentiation, and neuromodulator-induced potentiation.
Collapse
Affiliation(s)
- Melissa Silva
- Université de Paris, SPPIN-Saints Pères Paris Institute for the Neurosciences, CNRS, F-75006 Paris, France
| | - Van Tran
- Université de Paris, SPPIN-Saints Pères Paris Institute for the Neurosciences, CNRS, F-75006 Paris, France
| | - Alain Marty
- Université de Paris, SPPIN-Saints Pères Paris Institute for the Neurosciences, CNRS, F-75006 Paris, France.
| |
Collapse
|
46
|
Emperador-Melero J, Wong MY, Wang SSH, de Nola G, Nyitrai H, Kirchhausen T, Kaeser PS. PKC-phosphorylation of Liprin-α3 triggers phase separation and controls presynaptic active zone structure. Nat Commun 2021; 12:3057. [PMID: 34031393 PMCID: PMC8144191 DOI: 10.1038/s41467-021-23116-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/16/2021] [Indexed: 01/24/2023] Open
Abstract
The active zone of a presynaptic nerve terminal defines sites for neurotransmitter release. Its protein machinery may be organized through liquid-liquid phase separation, a mechanism for the formation of membrane-less subcellular compartments. Here, we show that the active zone protein Liprin-α3 rapidly and reversibly undergoes phase separation in transfected HEK293T cells. Condensate formation is triggered by Liprin-α3 PKC-phosphorylation at serine-760, and RIM and Munc13 are co-recruited into membrane-attached condensates. Phospho-specific antibodies establish phosphorylation of Liprin-α3 serine-760 in transfected cells and mouse brain tissue. In primary hippocampal neurons of newly generated Liprin-α2/α3 double knockout mice, synaptic levels of RIM and Munc13 are reduced and the pool of releasable vesicles is decreased. Re-expression of Liprin-α3 restored these presynaptic defects, while mutating the Liprin-α3 phosphorylation site to abolish phase condensation prevented this rescue. Finally, PKC activation in these neurons acutely increased RIM, Munc13 and neurotransmitter release, which depended on the presence of phosphorylatable Liprin-α3. Our findings indicate that PKC-mediated phosphorylation of Liprin-α3 triggers its phase separation and modulates active zone structure and function.
Collapse
Affiliation(s)
| | - Man Yan Wong
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Hajnalka Nyitrai
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,VIB-KU Leuven Center for Brain and Disease Research, Campus Gasthuisberg, Leuven, Belgium
| | - Tom Kirchhausen
- Departments of Cell Biology and Pediatrics, Harvard Medical School and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
A Trio of Active Zone Proteins Comprised of RIM-BPs, RIMs, and Munc13s Governs Neurotransmitter Release. Cell Rep 2021; 32:107960. [PMID: 32755572 DOI: 10.1016/j.celrep.2020.107960] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/01/2020] [Accepted: 07/02/2020] [Indexed: 11/21/2022] Open
Abstract
At the presynaptic active zone, action-potential-triggered neurotransmitter release requires that fusion-competent synaptic vesicles are placed next to Ca2+ channels. The active zone resident proteins RIM, RBP, and Munc13 are essential contributors for vesicle priming and Ca2+-channel recruitment. Although the individual contributions of these scaffolds have been extensively studied, their respective functions in neurotransmission are still incompletely understood. Here, we analyze the functional interactions of RIMs, RBPs, and Munc13s at the genetic, molecular, functional, and ultrastructural levels in a mammalian synapse. We find that RBP, together with Munc13, promotes vesicle priming at the expense of RBP's role in recruiting presynaptic Ca2+ channels, suggesting that the support of RBP for vesicle priming and Ca2+-secretion coupling is mutually exclusive. Our results demonstrate that the functional interaction of RIM, RBP, and Munc13 is more profound than previously envisioned, acting as a functional trio that govern basic and short-term plasticity properties of neurotransmission.
Collapse
|
48
|
Karlocai MR, Heredi J, Benedek T, Holderith N, Lorincz A, Nusser Z. Variability in the Munc13-1 content of excitatory release sites. eLife 2021; 10:67468. [PMID: 33904397 PMCID: PMC8116053 DOI: 10.7554/elife.67468] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
The molecular mechanisms underlying the diversity of cortical glutamatergic synapses are still incompletely understood. Here, we tested the hypothesis that presynaptic active zones (AZs) are constructed from molecularly uniform, independent release sites (RSs), the number of which scales linearly with the AZ size. Paired recordings between hippocampal CA1 pyramidal cells and fast-spiking interneurons in acute slices from adult mice followed by quantal analysis demonstrate large variability in the number of RSs (N) at these connections. High-resolution molecular analysis of functionally characterized synapses reveals variability in the content of one of the key vesicle priming factors – Munc13-1 – in AZs that possess the same N. Replica immunolabeling also shows a threefold variability in the total Munc13-1 content of AZs of identical size and a fourfold variability in the size and density of Munc13-1 clusters within the AZs. Our results provide evidence for quantitative molecular heterogeneity of RSs and support a model in which the AZ is built up from variable numbers of molecularly heterogeneous, but independent RSs.
Collapse
Affiliation(s)
- Maria Rita Karlocai
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Judit Heredi
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Tünde Benedek
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Noemi Holderith
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Andrea Lorincz
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
49
|
Choquet D, Sainlos M, Sibarita JB. Advanced imaging and labelling methods to decipher brain cell organization and function. Nat Rev Neurosci 2021; 22:237-255. [PMID: 33712727 DOI: 10.1038/s41583-021-00441-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2021] [Indexed: 01/31/2023]
Abstract
The brain is arguably the most complex organ. The branched and extended morphology of nerve cells, their subcellular complexity, the multiplicity of brain cell types as well as their intricate connectivity and the scattering properties of brain tissue present formidable challenges to the understanding of brain function. Neuroscientists have often been at the forefront of technological and methodological developments to overcome these hurdles to visualize, quantify and modify cell and network properties. Over the last few decades, the development of advanced imaging methods has revolutionized our approach to explore the brain. Super-resolution microscopy and tissue imaging approaches have recently exploded. These instrumentation-based innovations have occurred in parallel with the development of new molecular approaches to label protein targets, to evolve new biosensors and to target them to appropriate cell types or subcellular compartments. We review the latest developments for labelling and functionalizing proteins with small localization and functionalized reporters. We present how these molecular tools are combined with the development of a wide variety of imaging methods that break either the diffraction barrier or the tissue penetration depth limits. We put these developments in perspective to emphasize how they will enable step changes in our understanding of the brain.
Collapse
Affiliation(s)
- Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France. .,University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, Bordeaux, France.
| | - Matthieu Sainlos
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| | - Jean-Baptiste Sibarita
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
50
|
Yang X, Annaert W. The Nanoscopic Organization of Synapse Structures: A Common Basis for Cell Communication. MEMBRANES 2021; 11:248. [PMID: 33808285 PMCID: PMC8065904 DOI: 10.3390/membranes11040248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 11/16/2022]
Abstract
Synapse structures, including neuronal and immunological synapses, can be seen as the plasma membrane contact sites between two individual cells where information is transmitted from one cell to the other. The distance between the two plasma membranes is only a few tens of nanometers, but these areas are densely populated with functionally different proteins, including adhesion proteins, receptors, and transporters. The narrow space between the two plasma membranes has been a barrier for resolving the synaptic architecture due to the diffraction limit in conventional microscopy (~250 nm). Various advanced super-resolution microscopy techniques, such as stimulated emission depletion (STED), structured illumination microscopy (SIM), and single-molecule localization microscopy (SMLM), bypass the diffraction limit and provide a sub-diffraction-limit resolving power, ranging from 10 to 100 nm. The studies using super-resolution microscopy have revealed unprecedented details of the nanoscopic organization and dynamics of synaptic molecules. In general, most synaptic proteins appear to be heterogeneously distributed and form nanodomains at the membranes. These nanodomains are dynamic functional units, playing important roles in mediating signal transmission through synapses. Herein, we discuss our current knowledge on the super-resolution nanoscopic architecture of synapses and their functional implications, with a particular focus on the neuronal synapses and immune synapses.
Collapse
Affiliation(s)
| | - Wim Annaert
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Gasthuisberg, B-3000 Leuven, Belgium;
| |
Collapse
|