1
|
Murphy FH, Abramian A, Klaassen RV, Koopmans F, Persoon CM, Smit AB, Toonen RF, Verhage M. RIM and MUNC13 membrane-binding domains are essential for neuropeptide secretion. J Cell Biol 2025; 224:e202409196. [PMID: 40353777 PMCID: PMC12077229 DOI: 10.1083/jcb.202409196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/18/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
Neurons release neurotransmitters from synaptic vesicles (SVs) and neuropeptides from dense-core vesicles (DCVs). The presynaptic proteins RIM and MUNC13 play key roles in both pathways. It remains unclear how DCVs are targeted to release sites and whether RIM and MUNC13 are involved in this process. Here, we show that three membrane-binding domains in RIM and MUNC13 regulate DCV exocytosis differently from SV exocytosis. Using neuropeptide secretion assays with single-vesicle resolution and peptidomics analysis of endogenous neuropeptide release in MUNC13/RIM null neurons, we demonstrate that MUNC13 is essential for DCV exocytosis. The RIM N terminus prevents MUNC13 degradation via the proteasome, and inhibiting proteasomal degradation partially rescues DCV exocytosis in RIM's absence. Unlike SV exocytosis, the PIP2-binding RIM C2B domain and MUNC13 C1-C2B polybasic face are redundant for DCV exocytosis, while the lipid-binding MUNC13 C2C domain is crucial. These results show that RIM and MUNC13 synergistically regulate DCV exocytosis through membrane interactions and reveal new mechanistic differences between SV and DCV exocytosis.
Collapse
Affiliation(s)
- Fiona H. Murphy
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| | - Adlin Abramian
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| | - Remco V. Klaassen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| | - Frank Koopmans
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| | - Claudia M. Persoon
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - August B. Smit
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| | - Ruud F. Toonen
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| | - Matthijs Verhage
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
2
|
Sun R, Allen JP, Mao Z, Wilson L, Haider M, Alten B, Zhou Z, Wang X, Zhou Q. The postsynaptic density in excitatory synapses is composed of clustered, heterogeneous nanoblocks. J Cell Biol 2025; 224:e202406133. [PMID: 40145863 PMCID: PMC11948668 DOI: 10.1083/jcb.202406133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
The nanoscale organization of proteins within synapses is critical for maintaining and regulating synaptic transmission and plasticity. Here, we used cryo-electron tomography (cryo-ET) to directly visualize the three-dimensional architecture and supramolecular organization of postsynaptic components in both synaptosomes and synapses from cultured neurons. Cryo-ET revealed that postsynaptic density (PSD) is composed of membrane-associated nanoblocks of various sizes. Subtomogram averaging from synaptosomes showed two types (type A and B) of postsynaptic receptor-like particles at resolutions of 24 and 26 Å, respectively. Furthermore, our analysis suggested that potential presynaptic release sites are closer to nanoblocks with type A/B receptor-like particles than to nanoblocks without type A/B receptor-like particles. The results of this study provide a more comprehensive understanding of synaptic ultrastructure and suggest that PSD is composed of clustering of various nanoblocks. These nanoblocks are heterogeneous in size, assembly, and distribution, which likely contribute to the dynamic nature of PSD in modulating synaptic strength.
Collapse
Affiliation(s)
- Rong Sun
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - James P. Allen
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Zhuqing Mao
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Liana Wilson
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Mariam Haider
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology Cryo-EM Facility, Vanderbilt University, Nashville, TN, USA
| | - Baris Alten
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Zimeng Zhou
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- School of Engineering, Vanderbilt University, Nashville, TN, USA
| | - Xinyi Wang
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Peabody College, Vanderbilt University, Nashville, TN, USA
| | - Qiangjun Zhou
- Department of Cell and Developmental Biology, Center for Structural Biology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
3
|
Lin KH, Ranjan M, Lipstein N, Brose N, Neher E, Taschenberger H. Number and relative abundance of synaptic vesicles in functionally distinct priming states determine synaptic strength and short-term plasticity. J Physiol 2025. [PMID: 40120134 DOI: 10.1113/jp286282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Heterogeneity in synaptic strength and short-term plasticity (STP) was characterized in post-hearing rat calyx of Held synapses at near-physiological external [Ca2+] under control conditions and after experimentally induced synaptic potentiation. Kinetic modelling was combined with non-negative tensor factorization (NTF) to separate changes in synaptic vesicle (SV) priming kinetics from those in SV fusion probability (pfu sion). Heterogeneous synaptic strength and STP under control conditions can be fully accounted for by assuming a uniform pfusion among calyx synapses yet profound synapse-to-synapse variation in the resting equilibrium of SVs in functionally distinct priming states. Although synaptic potentiation induced by either elevated resting [Ca2+]i, elevated external [Ca2+] or stimulation of the diacylglycerol (DAG) signalling pathway leads to seemingly similar changes, that is, stronger synapses with less facilitation and more pronounced depression, the underlying mechanisms are different. Specifically, synaptic potentiation induced by the DAG mimetic and Munc13/PKC activator phorbol 12,13-dibutyrate (PDBu) only moderately enhances pfusion but strongly increases the abundance of fusion-competent maturely primed SVs, demonstrating that the dynamic equilibrium of differentially primed SVs critically determines synaptic strength and STP. Activation of the DAG pathway not only stimulates priming at resting [Ca2+]i but further promotes SV pool replenishment at elevated [Ca2+]i following pool-depleting stimulus trains. A two-step priming and fusion scheme which recapitulates the sequential build-up of the molecular SV fusion machinery is capable of reproducing experimentally induced changes in synaptic strength and STP in numerical simulations with a small number of plausible model parameter changes. KEY POINTS: A relatively simple two-step synaptic vesicle (SV) priming and fusion scheme is capable of reproducing experimentally induced changes in synaptic strength and short-term plasticity with a small number of plausible parameter changes. The combination of non-negative tensor factorization (NTF)-decomposition analysis and state modelling allows one to separate experimentally induced changes in SV priming kinetics from those in SV fusion probability. A relatively low sensitivity of the SV priming equilibrium to changes in resting [Ca2+]i suggests that the amplitude of the 'effective' action potential (AP)-induced Ca2+ transient is quite large, likely representing contributions of global and local Ca2+ signals. Enhanced synaptic strength and stronger depression after stimulation of the diacylglycerol (DAG) signalling pathway is primarily caused by enhanced SV priming, leading to increased abundance of maturely primed SVs at rest with comparably small changes in SV fusion probability. Application of DAG mimetics enhances the Ca2+-dependent acceleration of SV priming causing a faster recovery of synaptic strength after pool-depleting stimuli.
Collapse
Affiliation(s)
- Kun-Han Lin
- Laboratory of Membrane Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mrinalini Ranjan
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, Göttingen, Germany
| | - Noa Lipstein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Erwin Neher
- Laboratory of Membrane Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
4
|
Petrovic A, Do TT, Fernández-Busnadiego R. New insights into the molecular architecture of neurons by cryo-electron tomography. Curr Opin Neurobiol 2025; 90:102939. [PMID: 39667254 DOI: 10.1016/j.conb.2024.102939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/10/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024]
Abstract
Cryo-electron tomography (cryo-ET) visualizes natively preserved cellular ultrastructure at molecular resolution. Recent developments in sample preparation workflows and image processing tools enable growing applications of cryo-ET in cellular neurobiology. As such, cryo-ET is beginning to unravel the in situ macromolecular organization of neurons using samples of increasing complexity. Here, we highlight advances in cryo-ET technology and review its recent use to study neuronal architecture and its alterations under disease conditions.
Collapse
Affiliation(s)
- Arsen Petrovic
- University Medical Center Göttingen, Institute for Neuropathology, Göttingen, 37077, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, 37077, Germany.
| | - Thanh Thao Do
- University Medical Center Göttingen, Institute for Neuropathology, Göttingen, 37077, Germany
| | - Rubén Fernández-Busnadiego
- University Medical Center Göttingen, Institute for Neuropathology, Göttingen, 37077, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, 37077, Germany; Faculty of Physics, University of Göttingen, Göttingen, 37077, Germany.
| |
Collapse
|
5
|
Berns MMM, Yildiz M, Winkelmann S, Walter AM. Independently engaging protein tethers of different length enhance synaptic vesicle trafficking to the plasma membrane. J Physiol 2025. [PMID: 39808523 DOI: 10.1113/jp286651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Synaptic vesicle (SV) trafficking toward the plasma membrane (PM) and subsequent SV maturation are essential for neurotransmitter release. These processes, including SV docking and priming, are co-ordinated by various proteins, such as SNAREs, Munc13 and synaptotagmin (Syt), which connect (tether) the SV to the PM. Here, we investigated how tethers of varying lengths mediate SV docking using a simplified mathematical model. The heights of the three tether types, as estimated from the structures of the SNARE complex, Munc13 and Syt, defined the SV-PM distance ranges for tether formation. Geometric considerations linked SV-PM distances to the probability and rate of tether formation. We assumed that SV tethering constrains SV motility and that multiple tethers are associated by independent interactions. The model predicted that forming multiple tethers favours shorter SV-PM distances. Although tethers acted independently in the model, their geometrical properties often caused their sequential assembly, from longer ones (Munc13/Syt), which accelerated SV movement towards the PM, to shorter ones (SNAREs), which stabilized PM-proximal SVs. Modifying tether lengths or numbers affected SV trafficking. The independent implementation of tethering proteins enabled their selective removal to mimic gene knockout (KO) situations. This showed that simulated SV-PM distance distributions qualitatively aligned with published electron microscopy studies upon removal of SNARE and Syt tethers, whereas Munc13 KO data were best approximated when assuming additional disruption of SNARE tethers. Thus, although salient features of SV docking can be accounted for by independent tethering alone, our results suggest that functional tether interactions not yet featured in our model are crucial for biological function. KEY POINTS: A mathematical model describing the role of synaptic protein tethers to localize transmitter-containing vesicles is developed based on geometrical considerations and structural information of synaptotagmin, Munc13 and SNARE proteins. Vesicle movement, along with tether association and dissociation, are modelled as stochastic processes, with tethers functioning independently of each other. Multiple tethers cooperate to recruit vesicles to the plasma membrane and keep them there: Munc13 and Syt as the longer tethers accelerate the movement towards the membrane, whereas short SNARE tethers stabilize them there. Model predictions for situations in which individual tethers are removed agree with the results from experimental studies upon gene knockout. Changing tether length or copy numbers affects vesicle trafficking and steady-state distributions.
Collapse
Affiliation(s)
- Manon M M Berns
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Alexander M Walter
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Reshetniak S, Bogaciu CA, Bonn S, Brose N, Cooper BH, D'Este E, Fauth M, Fernández-Busnadiego R, Fiosins M, Fischer A, Georgiev SV, Jakobs S, Klumpp S, Köster S, Lange F, Lipstein N, Macarrón-Palacios V, Milovanovic D, Moser T, Müller M, Opazo F, Outeiro TF, Pape C, Priesemann V, Rehling P, Salditt T, Schlüter O, Simeth N, Steinem C, Tchumatchenko T, Tetzlaff C, Tirard M, Urlaub H, Wichmann C, Wolf F, Rizzoli SO. The synaptic vesicle cluster as a controller of pre- and postsynaptic structure and function. J Physiol 2024. [PMID: 39367860 DOI: 10.1113/jp286400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024] Open
Abstract
The synaptic vesicle cluster (SVC) is an essential component of chemical synapses, which provides neurotransmitter-loaded vesicles during synaptic activity, at the same time as also controlling the local concentrations of numerous exo- and endocytosis cofactors. In addition, the SVC hosts molecules that participate in other aspects of synaptic function, from cytoskeletal components to adhesion proteins, and affects the location and function of organelles such as mitochondria and the endoplasmic reticulum. We argue here that these features extend the functional involvement of the SVC in synapse formation, signalling and plasticity, as well as synapse stabilization and metabolism. We also propose that changes in the size of the SVC coalesce with changes in the postsynaptic compartment, supporting the interplay between pre- and postsynaptic dynamics. Thereby, the SVC could be seen as an 'all-in-one' regulator of synaptic structure and function, which should be investigated in more detail, to reveal molecular mechanisms that control synaptic function and heterogeneity.
Collapse
Affiliation(s)
- Sofiia Reshetniak
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Cristian A Bogaciu
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Elisa D'Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Michael Fauth
- Georg-August-University Göttingen, Faculty of Physics, Institute for the Dynamics of Complex Systems, Friedrich-Hund-Platz 1, Göttingen, Germany
| | - Rubén Fernández-Busnadiego
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Maksims Fiosins
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - André Fischer
- German Center for Neurodegenerative Diseases, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Svilen V Georgiev
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Jakobs
- Research Group Structure and Dynamics of Mitochondria, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Stefan Klumpp
- Theoretical Biophysics Group, Institute for the Dynamics of Complex Systems, Georg-August University Göttingen, Göttingen, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Felix Lange
- Research Group Structure and Dynamics of Mitochondria, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Noa Lipstein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Marcus Müller
- Institute for Theoretical Physics, Georg-August University Göttingen, Göttingen, Germany
| | - Felipe Opazo
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Constantin Pape
- Institute of Computer Science, Georg-August University Göttingen, Göttingen, Germany
| | - Viola Priesemann
- Georg-August-University Göttingen, Faculty of Physics, Institute for the Dynamics of Complex Systems, Friedrich-Hund-Platz 1, Göttingen, Germany
- Max-Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institute for X-Ray Physics, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Oliver Schlüter
- Clinic for Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nadja Simeth
- Institute of Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
| | - Christian Tetzlaff
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Marilyn Tirard
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Carolin Wichmann
- Institute for Auditory Neuroscience University Medical Center Göttingen, Göttingen, Germany
- Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Fred Wolf
- Max-Planck-Institute for Dynamics and Self-Organization, 37077 Göttingen and Institute for Dynamics of Biological Networks, Georg-August University Göttingen, Göttingen, Germany
| | - Silvio O Rizzoli
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
7
|
Zheng T, Cai S. Recent technical advances in cellular cryo-electron tomography. Int J Biochem Cell Biol 2024; 175:106648. [PMID: 39181502 DOI: 10.1016/j.biocel.2024.106648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Understanding the in situ structure, organization, and interactions of macromolecules is essential for elucidating their functions and mechanisms of action. Cellular cryo-electron tomography (cryo-ET) is a cutting-edge technique that reveals in situ molecular-resolution architectures of macromolecules in their lifelike states. It also provides insights into the three-dimensional distribution of macromolecules and their spatial relationships with various subcellular structures. Thus, cellular cryo-ET bridges the gap between structural biology and cell biology. With rapid advancements, this technique achieved substantial improvements in throughput, automation, and resolution. This review presents the fundamental principles and methodologies of cellular cryo-ET, highlighting recent developments in sample preparation, data collection, and image processing. We also discuss emerging trends and potential future directions. As cellular cryo-ET continues to develop, it is set to play an increasingly vital role in structural cell biology.
Collapse
Affiliation(s)
- Tianyu Zheng
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shujun Cai
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
8
|
Held RG, Liang J, Brunger AT. Nanoscale architecture of synaptic vesicles and scaffolding complexes revealed by cryo-electron tomography. Proc Natl Acad Sci U S A 2024; 121:e2403136121. [PMID: 38923992 PMCID: PMC11228483 DOI: 10.1073/pnas.2403136121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The spatial distribution of proteins and their arrangement within the cellular ultrastructure regulates the opening of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in response to glutamate release at the synapse. Fluorescence microscopy imaging revealed that the postsynaptic density (PSD) and scaffolding proteins in the presynaptic active zone (AZ) align across the synapse to form a trans-synaptic "nanocolumn," but the relation to synaptic vesicle release sites is uncertain. Here, we employ focused-ion beam (FIB) milling and cryoelectron tomography to image synapses under near-native conditions. Improved image contrast, enabled by FIB milling, allows simultaneous visualization of supramolecular nanoclusters within the AZ and PSD and synaptic vesicles. Surprisingly, membrane-proximal synaptic vesicles, which fuse to release glutamate, are not preferentially aligned with AZ or PSD nanoclusters. These synaptic vesicles are linked to the membrane by peripheral protein densities, often consistent in size and shape with Munc13, as well as globular densities bridging the synaptic vesicle and plasma membrane, consistent with prefusion complexes of SNAREs, synaptotagmins, and complexin. Monte Carlo simulations of synaptic transmission events using biorealistic models guided by our tomograms predict that clustering AMPARs within PSD nanoclusters increases the variability of the postsynaptic response but not its average amplitude. Together, our data support a model in which synaptic strength is tuned at the level of single vesicles by the spatial relationship between scaffolding nanoclusters and single synaptic vesicle fusion sites.
Collapse
Affiliation(s)
- Richard G. Held
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Jiahao Liang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Axel T. Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA94305
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA94305
- Department of Structural Biology, Stanford University, Stanford, CA94305
- Department of Photon Science, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| |
Collapse
|
9
|
Mrestani A, Dannhäuser S, Pauli M, Kollmannsberger P, Hübsch M, Morris L, Langenhan T, Heckmann M, Paul MM. Nanoscaled RIM clustering at presynaptic active zones revealed by endogenous tagging. Life Sci Alliance 2023; 6:e202302021. [PMID: 37696575 PMCID: PMC10494931 DOI: 10.26508/lsa.202302021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
Chemical synaptic transmission involves neurotransmitter release from presynaptic active zones (AZs). The AZ protein Rab-3-interacting molecule (RIM) is important for normal Ca2+-triggered release. However, its precise localization within AZs of the glutamatergic neuromuscular junctions of Drosophila melanogaster remains elusive. We used CRISPR/Cas9-assisted genome engineering of the rim locus to incorporate small epitope tags for targeted super-resolution imaging. A V5-tag, derived from simian virus 5, and an HA-tag, derived from human influenza virus, were N-terminally fused to the RIM Zinc finger. Whereas both variants are expressed in co-localization with the core AZ scaffold Bruchpilot, electrophysiological characterization reveals that AP-evoked synaptic release is disturbed in rimV5-Znf but not in rimHA-Znf In addition, rimHA-Znf synapses show intact presynaptic homeostatic potentiation. Combining super-resolution localization microscopy and hierarchical clustering, we detect ∼10 RIMHA-Znf subclusters with ∼13 nm diameter per AZ that are compacted and increased in numbers in presynaptic homeostatic potentiation.
Collapse
Affiliation(s)
- 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
| | - Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Martin Pauli
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Martha Hübsch
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lydia Morris
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Tobias Langenhan
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, 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
|
10
|
Papantoniou C, Laugks U, Betzin J, Capitanio C, Ferrero JJ, Sánchez-Prieto J, Schoch S, Brose N, Baumeister W, Cooper BH, Imig C, Lučić V. Munc13- and SNAP25-dependent molecular bridges play a key role in synaptic vesicle priming. SCIENCE ADVANCES 2023; 9:eadf6222. [PMID: 37343100 PMCID: PMC10284560 DOI: 10.1126/sciadv.adf6222] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 05/17/2023] [Indexed: 06/23/2023]
Abstract
Synaptic vesicle tethering, priming, and neurotransmitter release require a coordinated action of multiple protein complexes. While physiological experiments, interaction data, and structural studies of purified systems were essential for our understanding of the function of the individual complexes involved, they cannot resolve how the actions of individual complexes integrate. We used cryo-electron tomography to simultaneously image multiple presynaptic protein complexes and lipids at molecular resolution in their native composition, conformation, and environment. Our detailed morphological characterization suggests that sequential synaptic vesicle states precede neurotransmitter release, where Munc13-comprising bridges localize vesicles <10 nanometers and soluble N-ethylmaleimide-sensitive factor attachment protein 25-comprising bridges <5 nanometers from the plasma membrane, the latter constituting a molecularly primed state. Munc13 activation supports the transition to the primed state via vesicle bridges to plasma membrane (tethers), while protein kinase C promotes the same transition by reducing vesicle interlinking. These findings exemplify a cellular function performed by an extended assembly comprising multiple molecularly diverse complexes.
Collapse
Affiliation(s)
- Christos Papantoniou
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Ulrike Laugks
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Julia Betzin
- Department of Neuropathology, University Hospital of Bonn, 53127 Bonn, Germany
| | - Cristina Capitanio
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - José Javier Ferrero
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Susanne Schoch
- Department of Neuropathology, University Hospital of Bonn, 53127 Bonn, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Benjamin H. Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Multidisciplinary Sciences, City Campus, 37075 Göttingen, Germany
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Vladan Lučić
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| |
Collapse
|
11
|
Radecke J, Seeger R, Kádková A, Laugks U, Khosrozadeh A, Goldie KN, Lučić V, Sørensen JB, Zuber B. Morphofunctional changes at the active zone during synaptic vesicle exocytosis. EMBO Rep 2023; 24:e55719. [PMID: 36876590 PMCID: PMC10157379 DOI: 10.15252/embr.202255719] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/30/2023] [Accepted: 02/16/2023] [Indexed: 03/07/2023] Open
Abstract
Synaptic vesicle (SV) fusion with the plasma membrane (PM) proceeds through intermediate steps that remain poorly resolved. The effect of persistent high or low exocytosis activity on intermediate steps remains unknown. Using spray-mixing plunge-freezing cryo-electron tomography we observe events following synaptic stimulation at nanometer resolution in near-native samples. Our data suggest that during the stage that immediately follows stimulation, termed early fusion, PM and SV membrane curvature changes to establish a point contact. The next stage-late fusion-shows fusion pore opening and SV collapse. During early fusion, proximal tethered SVs form additional tethers with the PM and increase the inter-SV connector number. In the late-fusion stage, PM-proximal SVs lose their interconnections, allowing them to move toward the PM. Two SNAP-25 mutations, one arresting and one disinhibiting spontaneous release, cause connector loss. The disinhibiting mutation causes loss of membrane-proximal multiple-tethered SVs. Overall, tether formation and connector dissolution are triggered by stimulation and respond to spontaneous fusion rate manipulation. These morphological observations likely correspond to SV transition from one functional pool to another.
Collapse
Affiliation(s)
- Julika Radecke
- Institute of AnatomyUniversity of BernBernSwitzerland
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
- Diamond Light Source LtdDidcotUK
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Raphaela Seeger
- Institute of AnatomyUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Anna Kádková
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Ulrike Laugks
- Max‐Planck‐Institute of BiochemistryMartinsriedGermany
| | - Amin Khosrozadeh
- Institute of AnatomyUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | | | - Vladan Lučić
- Max‐Planck‐Institute of BiochemistryMartinsriedGermany
| | - Jakob B Sørensen
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Benoît Zuber
- Institute of AnatomyUniversity of BernBernSwitzerland
| |
Collapse
|
12
|
Martín R, Suárez-Pinilla AS, García-Font N, Laguna-Luque ML, López-Ramos JC, Oset-Gasque MJ, Gruart A, Delgado-García JM, Torres M, Sánchez-Prieto J. The activation of mGluR4 rescues parallel fiber synaptic transmission and LTP, motor learning and social behavior in a mouse model of Fragile X Syndrome. Mol Autism 2023; 14:14. [PMID: 37029391 PMCID: PMC10082511 DOI: 10.1186/s13229-023-00547-4] [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: 09/12/2022] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS), the most common inherited intellectual disability, is caused by the loss of expression of the Fragile X Messenger Ribonucleoprotein (FMRP). FMRP is an RNA-binding protein that negatively regulates the expression of many postsynaptic as well as presynaptic proteins involved in action potential properties, calcium homeostasis and neurotransmitter release. FXS patients and mice lacking FMRP suffer from multiple behavioral alterations, including deficits in motor learning for which there is currently no specific treatment. METHODS We performed electron microscopy, whole-cell patch-clamp electrophysiology and behavioral experiments to characterise the synaptic mechanisms underlying the motor learning deficits observed in Fmr1KO mice and the therapeutic potential of positive allosteric modulator of mGluR4. RESULTS We found that enhanced synaptic vesicle docking of cerebellar parallel fiber to Purkinje cell Fmr1KO synapses was associated with enhanced asynchronous release, which not only prevents further potentiation, but it also compromises presynaptic parallel fiber long-term potentiation (PF-LTP) mediated by β adrenergic receptors. A reduction in extracellular Ca2+ concentration restored the readily releasable pool (RRP) size, basal synaptic transmission, β adrenergic receptor-mediated potentiation, and PF-LTP. Interestingly, VU 0155041, a selective positive allosteric modulator of mGluR4, also restored both the RRP size and PF-LTP in mice of either sex. Moreover, when injected into Fmr1KO male mice, VU 0155041 improved motor learning in skilled reaching, classical eyeblink conditioning and vestibuloocular reflex (VOR) tests, as well as the social behavior alterations of these mice. LIMITATIONS We cannot rule out that the activation of mGluR4s via systemic administration of VU0155041 can also affect other brain regions. Further studies are needed to stablish the effect of a specific activation of mGluR4 in cerebellar granule cells. CONCLUSIONS Our study shows that an increase in synaptic vesicles, SV, docking may cause the loss of PF-LTP and motor learning and social deficits of Fmr1KO mice and that the reversal of these changes by pharmacological activation of mGluR4 may offer therapeutic relief for motor learning and social deficits in FXS.
Collapse
Affiliation(s)
- Ricardo Martín
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Instituto Universitario de Investigación en Neuroquímica, 28040, Madrid, Spain.
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain.
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense, 28040, Madrid, Spain.
| | - Alberto Samuel Suárez-Pinilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Instituto Universitario de Investigación en Neuroquímica, 28040, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Nuria García-Font
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Instituto Universitario de Investigación en Neuroquímica, 28040, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
- Centre for Discovery Brain Sciences and Simon Initiative for Developing Brain, University of Edinburgh, Edinburgh, EH89JZ, UK
| | | | - Juan C López-Ramos
- Division de Neurociencias, Universidad Pablo de Olavide, 41013, Sevilla, Spain
| | - María Jesús Oset-Gasque
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
- Departamento de Bioquímica, Facultad de Farmacia, Universidad Complutense, Instituto Universitario Investigación en Neuroquímica, 28040, Madrid, Spain
| | - Agnes Gruart
- Division de Neurociencias, Universidad Pablo de Olavide, 41013, Sevilla, Spain
| | | | - Magdalena Torres
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Instituto Universitario de Investigación en Neuroquímica, 28040, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Instituto Universitario de Investigación en Neuroquímica, 28040, Madrid, Spain.
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain.
| |
Collapse
|
13
|
Digitalizing neuronal synapses with cryo-electron tomography and correlative microscopy. Curr Opin Neurobiol 2022; 76:102595. [DOI: 10.1016/j.conb.2022.102595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/22/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
|
14
|
Lichter K, Paul MM, Pauli M, Schoch S, Kollmannsberger P, Stigloher C, Heckmann M, Sirén AL. Ultrastructural analysis of wild-type and RIM1α knockout active zones in a large cortical synapse. Cell Rep 2022; 40:111382. [PMID: 36130490 DOI: 10.1016/j.celrep.2022.111382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/14/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022] Open
Abstract
Rab3A-interacting molecule (RIM) is crucial for fast Ca2+-triggered synaptic vesicle (SV) release in presynaptic active zones (AZs). We investigated hippocampal giant mossy fiber bouton (MFB) AZ architecture in 3D using electron tomography of rapid cryo-immobilized acute brain slices in RIM1α-/- and wild-type mice. In RIM1α-/-, AZs are larger with increased synaptic cleft widths and a 3-fold reduced number of tightly docked SVs (0-2 nm). The distance of tightly docked SVs to the AZ center is increased from 110 to 195 nm, and the width of their electron-dense material between outer SV membrane and AZ membrane is reduced. Furthermore, the SV pool in RIM1α-/- is more heterogeneous. Thus, RIM1α, besides its role in tight SV docking, is crucial for synaptic architecture and vesicle pool organization in MFBs.
Collapse
Affiliation(s)
- Katharina Lichter
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany; Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Mila Marie Paul
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Martin Pauli
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
| | - Susanne Schoch
- Department of Neuropathology and Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius-Maximilians-University Würzburg, 97074 Würzburg, Germany
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Würzburg, 97074 Würzburg, Germany.
| | - Manfred Heckmann
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany.
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany; Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany.
| |
Collapse
|
15
|
Chen X, Kuner T, Blanpied TA. Editorial: Quantifying and controlling the nano-architecture of neuronal synapses. Front Synaptic Neurosci 2022; 14:1024073. [PMID: 36160915 PMCID: PMC9491271 DOI: 10.3389/fnsyn.2022.1024073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Xiaobing Chen
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Xiaobing Chen
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Thomas Kuner
| | - Thomas A. Blanpied
- School of Medicine, University of Maryland, Baltimore, MD, United States
- Thomas A. Blanpied
| |
Collapse
|
16
|
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
|
17
|
Wichmann C, Kuner T. Heterogeneity of glutamatergic synapses: cellular mechanisms and network consequences. Physiol Rev 2022; 102:269-318. [PMID: 34727002 DOI: 10.1152/physrev.00039.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chemical synapses are commonly known as a structurally and functionally highly diverse class of cell-cell contacts specialized to mediate communication between neurons. They represent the smallest "computational" unit of the brain and are typically divided into excitatory and inhibitory as well as modulatory categories. These categories are subdivided into diverse types, each representing a different structure-function repertoire that in turn are thought to endow neuronal networks with distinct computational properties. The diversity of structure and function found among a given category of synapses is referred to as heterogeneity. The main building blocks for this heterogeneity are synaptic vesicles, the active zone, the synaptic cleft, the postsynaptic density, and glial processes associated with the synapse. Each of these five structural modules entails a distinct repertoire of functions, and their combination specifies the range of functional heterogeneity at mammalian excitatory synapses, which are the focus of this review. We describe synapse heterogeneity that is manifested on different levels of complexity ranging from the cellular morphology of the pre- and postsynaptic cells toward the expression of different protein isoforms at individual release sites. We attempt to define the range of structural building blocks that are used to vary the basic functional repertoire of excitatory synaptic contacts and discuss sources and general mechanisms of synapse heterogeneity. Finally, we explore the possible impact of synapse heterogeneity on neuronal network function.
Collapse
Affiliation(s)
- Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg, Germany
| |
Collapse
|
18
|
Chakrabarti R, Jaime Tobón LM, Slitin L, Redondo Canales M, Hoch G, Slashcheva M, Fritsch E, Bodensiek K, Özçete ÖD, Gültas M, Michanski S, Opazo F, Neef J, Pangrsic T, Moser T, Wichmann C. Optogenetics and electron tomography for structure-function analysis of cochlear ribbon synapses. eLife 2022; 11:79494. [PMID: 36562477 PMCID: PMC9908081 DOI: 10.7554/elife.79494] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Ribbon synapses of cochlear inner hair cells (IHCs) are specialized to indefatigably transmit sound information at high rates. To understand the underlying mechanisms, structure-function analysis of the active zone (AZ) of these synapses is essential. Previous electron microscopy studies of synaptic vesicle (SV) dynamics at the IHC AZ used potassium stimulation, which limited the temporal resolution to minutes. Here, we established optogenetic IHC stimulation followed by quick freezing within milliseconds and electron tomography to study the ultrastructure of functional synapse states with good temporal resolution in mice. We characterized optogenetic IHC stimulation by patch-clamp recordings from IHCs and postsynaptic boutons revealing robust IHC depolarization and neurotransmitter release. Ultrastructurally, the number of docked SVs increased upon short (17-25 ms) and long (48-76 ms) light stimulation paradigms. We did not observe enlarged SVs or other morphological correlates of homotypic fusion events. Our results indicate a rapid recruitment of SVs to the docked state upon stimulation and suggest that univesicular release prevails as the quantal mechanism of exocytosis at IHC ribbon synapses.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Lina María Jaime Tobón
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Loujin Slitin
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Magdalena Redondo Canales
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Gerhard Hoch
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Marina Slashcheva
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of GöttingenGöttingenGermany
| | - Elisabeth Fritsch
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of GöttingenGöttingenGermany
| | - Kai Bodensiek
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany
| | - Özge Demet Özçete
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Mehmet Gültas
- Faculty of Agriculture, South Westphalia University of Applied SciencesSoestGermany
| | - Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,NanoTag Biotechnologies GmbHGöttingenGermany,Institute of Neuro- and Sensory Physiology, University Medical Center GöttingenGöttingenGermany
| | - Jakob Neef
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Tina Pangrsic
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany,Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany
| | - Tobias Moser
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany
| |
Collapse
|
19
|
Zuber B, Lučić V. Neurons as a model system for cryo-electron tomography. J Struct Biol X 2022; 6:100067. [PMID: 35310407 PMCID: PMC8924422 DOI: 10.1016/j.yjsbx.2022.100067] [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] [Indexed: 11/24/2022] Open
Abstract
Cryo-ET imaging of neurons is a versatile system for cell biology in situ. Structural and spatial localization analysis yields new insights into synaptic transmission. The synapse provides a rich environment for the development of image processing tools.
Cryo-electron tomography (Cryo-ET) provides unique opportunities to image cellular components at high resolution in their native state and environment. While many different cell types were investigated by cryo-ET, here we review application to neurons. We show that neurons are a versatile system that can be used to investigate general cellular components such as the cytoskeleton and membrane-bound organelles, in addition to neuron-specific processes such as synaptic transmission. Furthermore, the synapse provides a rich environment for the development of cryo-ET image processing tools suitable to elucidate the functional and spatial organization of compositionally and morphologically heterogeneous macromolecular complexes involved in biochemical signaling cascades, within their native, crowded cellular environments.
Collapse
|
20
|
Mochida S. Stable and Flexible Synaptic Transmission Controlled by the Active Zone Protein Interactions. Int J Mol Sci 2021; 22:ijms222111775. [PMID: 34769208 PMCID: PMC8583982 DOI: 10.3390/ijms222111775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022] Open
Abstract
An action potential triggers neurotransmitter release from synaptic vesicles docking to a specialized release site of the presynaptic plasma membrane, the active zone. The active zone is a highly organized structure with proteins that serves as a platform for synaptic vesicle exocytosis, mediated by SNAREs complex and Ca2+ sensor proteins, within a sub-millisecond opening of nearby Ca2+ channels with the membrane depolarization. In response to incoming neuronal signals, each active zone protein plays a role in the release-ready site replenishment with synaptic vesicles for sustainable synaptic transmission. The active zone release apparatus provides a possible link between neuronal activity and plasticity. This review summarizes the mostly physiological role of active zone protein interactions that control synaptic strength, presynaptic short-term plasticity, and homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Shortened tethering filaments stabilize presynaptic vesicles in support of elevated release probability during LTP in rat hippocampus. Proc Natl Acad Sci U S A 2021; 118:2018653118. [PMID: 33875591 DOI: 10.1073/pnas.2018653118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Long-term potentiation (LTP) is a cellular mechanism of learning and memory that results in a sustained increase in the probability of vesicular release of neurotransmitter. However, previous work in hippocampal area CA1 of the adult rat revealed that the total number of vesicles per synapse decreases following LTP, seemingly inconsistent with the elevated release probability. Here, electron-microscopic tomography (EMT) was used to assess whether changes in vesicle density or structure of vesicle tethering filaments at the active zone might explain the enhanced release probability following LTP. The spatial relationship of vesicles to the active zone varies with functional status. Tightly docked vesicles contact the presynaptic membrane, have partially formed SNARE complexes, and are primed for release of neurotransmitter upon the next action potential. Loosely docked vesicles are located within 8 nm of the presynaptic membrane where SNARE complexes begin to form. Nondocked vesicles comprise recycling and reserve pools. Vesicles are tethered to the active zone via filaments composed of molecules engaged in docking and release processes. The density of tightly docked vesicles was increased 2 h following LTP compared to control stimulation, whereas the densities of loosely docked or nondocked vesicles congregating within 45 nm above the active zones were unchanged. The tethering filaments on all vesicles were shorter and their attachment sites shifted closer to the active zone. These findings suggest that tethering filaments stabilize more vesicles in the primed state. Such changes would facilitate the long-lasting increase in release probability following LTP.
Collapse
|
24
|
The vesicle cluster as a major organizer of synaptic composition in the short-term and long-term. Curr Opin Cell Biol 2021; 71:63-68. [PMID: 33706235 DOI: 10.1016/j.ceb.2021.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/22/2021] [Accepted: 02/04/2021] [Indexed: 01/29/2023]
Abstract
For decades, the synaptic vesicle cluster has been thought of as a storage space for synaptic vesicles, whose obvious function is to provide vesicles for the depolarization-induced release of neurotransmitters; however, reports over the last few years indicate that the synaptic vesicle cluster probably plays a much broader and more fundamental role in synaptic biology. Various experiments suggest that the cluster is able to regulate protein distribution and mobility in the synapse; moreover, it probably regulates cytoskeleton architecture, mediates the selective removal of synaptic components from the bouton, and controls the responses of the presynapse to plasticity. Here we discuss these features of the vesicle cluster and conclude that it serves as a key organizer of synaptic composition and dynamics.
Collapse
|
25
|
Hintze A, Gültas M, Semmelhack EA, Wichmann C. Ultrastructural maturation of the endbulb of Held active zones comparing wild-type and otoferlin-deficient mice. iScience 2021; 24:102282. [PMID: 33851098 PMCID: PMC8022229 DOI: 10.1016/j.isci.2021.102282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/18/2021] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Endbulbs of Held are located in the anteroventral cochlear nucleus and present the first central synapses of the auditory pathway. During development, endbulbs mature functionally to enable rapid and powerful synaptic transmission with high temporal precision. This process is accompanied by morphological changes of endbulb terminals. Loss of the hair cell-specific protein otoferlin (Otof) abolishes neurotransmission in the cochlea and results in the smaller endbulb of Held terminals. Thus, peripheral hearing impairment likely also leads to alterations in the morphological synaptic vesicle (SV) pool size at individual endbulb of Held active zones (AZs). Here, we investigated endbulb AZs in pre-hearing, young, and adult wild-type and Otof−/− mice. During maturation, SV numbers at endbulb AZs increased in wild-type mice but were found to be reduced in Otof−/− mice. The SV population at a distance of 0–15 nm was most strongly affected. Finally, overall SV diameters decreased in Otof−/− animals during maturation. Maturation of wt endbulb of Held active zones leads to more synaptic vesicles At endbulbs of otoferlin knockout mice, synaptic vesicles decline with age Mainly two distinct synaptic vesicle populations are affected Synaptic vesicles sizes are reduced in six-month-old otoferlin knockout animals
Collapse
Affiliation(s)
- Anika Hintze
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.,Collaborative Research Center 1286, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Sciences, Georg-August-University Göttingen, Göttingen, Germany
| | - Esther A Semmelhack
- Developmental, Neural, and Behavioral Biology MSc/PhD Program, University of Göttingen, Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.,Collaborative Research Center 1286, University of Göttingen, Göttingen, Germany
| |
Collapse
|
26
|
Martinez-Sanchez A, Laugks U, Kochovski Z, Papantoniou C, Zinzula L, Baumeister W, Lučić V. Trans-synaptic assemblies link synaptic vesicles and neuroreceptors. SCIENCE ADVANCES 2021; 7:7/10/eabe6204. [PMID: 33674312 PMCID: PMC7935360 DOI: 10.1126/sciadv.abe6204] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/22/2021] [Indexed: 05/03/2023]
Abstract
Synaptic transmission is characterized by fast, tightly coupled processes and complex signaling pathways that require a precise protein organization, such as the previously reported nanodomain colocalization of pre- and postsynaptic proteins. Here, we used cryo-electron tomography to visualize synaptic complexes together with their native environment comprising interacting proteins and lipids on a 2- to 4-nm scale. Using template-free detection and classification, we showed that tripartite trans-synaptic assemblies (subcolumns) link synaptic vesicles to postsynaptic receptors and established that a particular displacement between directly interacting complexes characterizes subcolumns. Furthermore, we obtained de novo average structures of ionotropic glutamate receptors in their physiological composition, embedded in plasma membrane. These data support the hypothesis that synaptic function is carried by precisely organized trans-synaptic units. It provides a framework for further exploration of synaptic and other large molecular assemblies that link different cells or cellular regions and may require weak or transient interactions to exert their function.
Collapse
Affiliation(s)
- Antonio Martinez-Sanchez
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
- Department of Computer Sciences, Faculty of Sciences, University of Oviedo, Federico Garcia Lorca 18, 33007, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avenida Hospital Universitario s/n, 33011 Oviedo, Spain
- Institute of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Ulrike Laugks
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Zdravko Kochovski
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Christos Papantoniou
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Luca Zinzula
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Wolfgang Baumeister
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Vladan Lučić
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
27
|
Radhakrishnan A, Li X, Grushin K, Krishnakumar SS, Liu J, Rothman JE. Symmetrical arrangement of proteins under release-ready vesicles in presynaptic terminals. Proc Natl Acad Sci U S A 2021; 118:e2024029118. [PMID: 33468631 PMCID: PMC7865176 DOI: 10.1073/pnas.2024029118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Controlled release of neurotransmitters stored in synaptic vesicles (SVs) is a fundamental process that is central to all information processing in the brain. This relies on tight coupling of the SV fusion to action potential-evoked presynaptic Ca2+ influx. This Ca2+-evoked release occurs from a readily releasable pool (RRP) of SVs docked to the plasma membrane (PM). The protein components involved in initial SV docking/tethering and the subsequent priming reactions which make the SV release ready are known. Yet, the supramolecular architecture and sequence of molecular events underlying SV release are unclear. Here, we use cryoelectron tomography analysis in cultured hippocampal neurons to delineate the arrangement of the exocytosis machinery under docked SVs. Under native conditions, we find that vesicles are initially "tethered" to the PM by a variable number of protein densities (∼10 to 20 nm long) with no discernible organization. In contrast, we observe exactly six protein masses, each likely consisting of a single SNAREpin with its bound Synaptotagmins and Complexin, arranged symmetrically connecting the "primed" vesicles to the PM. Our data indicate that the fusion machinery is likely organized into a highly cooperative framework during the priming process which enables rapid SV fusion and neurotransmitter release following Ca2+ influx.
Collapse
Affiliation(s)
| | - Xia Li
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520
- Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06520
| | - Kirill Grushin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Shyam S Krishnakumar
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520;
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520;
- Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06520
| | - James E Rothman
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520;
| |
Collapse
|
28
|
Disentangling the Roles of RIM and Munc13 in Synaptic Vesicle Localization and Neurotransmission. J Neurosci 2020; 40:9372-9385. [PMID: 33139401 PMCID: PMC7724145 DOI: 10.1523/jneurosci.1922-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/22/2020] [Accepted: 10/15/2020] [Indexed: 11/21/2022] Open
Abstract
Efficient neurotransmitter release at the presynaptic terminal requires docking of synaptic vesicles to the active zone membrane and formation of fusion-competent synaptic vesicles near voltage-gated Ca2+ channels. Rab3-interacting molecule (RIM) is a critical active zone organizer, as it recruits Ca2+ channels and activates synaptic vesicle docking and priming via Munc13-1. However, our knowledge about Munc13-independent contributions of RIM to active zone functions is limited. To identify the functions that are solely mediated by RIM, we used genetic manipulations to control RIM and Munc13-1 activity in cultured hippocampal neurons from mice of either sex and compared synaptic ultrastructure and neurotransmission. We found that RIM modulates synaptic vesicle localization in the proximity of the active zone membrane independent of Munc13-1. In another step, both RIM and Munc13 mediate synaptic vesicle docking and priming. In addition, while the activity of both RIM and Munc13-1 is required for Ca2+-evoked release, RIM uniquely controls neurotransmitter release efficiency. However, activity-dependent augmentation of synaptic vesicle pool size relies exclusively on the action of Munc13s. Based on our results, we extend previous findings and propose a refined model in which RIM and Munc13-1 act in overlapping and independent stages of synaptic vesicle localization and release. SIGNIFICANCE STATEMENT The presynaptic active zone is composed of scaffolding proteins that functionally interact to localize synaptic vesicles to release sites, ensuring neurotransmission. Our current knowledge of the presynaptic active zone function relies on structure-function analysis, which has provided detailed information on the network of interactions and the impact of active zone proteins. Yet, the hierarchical, redundant, or independent cooperation of each active zone protein to synapse functions is not fully understood. Rab3-interacting molecule and Munc13 are the two key functionally interacting active zone proteins. Here, we dissected the distinct actions of Rab3-interacting molecule and Munc13-1 from both ultrastructural and physiological aspects. Our findings provide a more detailed view of how these two presynaptic proteins orchestrate their functions to achieve synaptic transmission.
Collapse
|
29
|
β-Adrenergic Receptors/Epac Signaling Increases the Size of the Readily Releasable Pool of Synaptic Vesicles Required for Parallel Fiber LTP. J Neurosci 2020; 40:8604-8617. [PMID: 33046543 DOI: 10.1523/jneurosci.0716-20.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 01/10/2023] Open
Abstract
The second messenger cAMP is an important determinant of synaptic plasticity that is associated with enhanced neurotransmitter release. Long-term potentiation (LTP) at parallel fiber (PF)-Purkinje cell (PC) synapses depends on a Ca2+-induced increase in presynaptic cAMP that is mediated by Ca2+-sensitive adenylyl cyclases. However, the upstream signaling and the downstream targets of cAMP involved in these events remain poorly understood. It is unclear whether cAMP generated by β-adrenergic receptors (βARs) is required for PF-PC LTP, although noradrenergic varicosities are apposed in PF-PC contacts. Guanine nucleotide exchange proteins directly activated by cAMP [Epac proteins (Epac 1-2)] are alternative cAMP targets to protein kinase A (PKA) and Epac2 is abundant in the cerebellum. However, whether Epac proteins participate in PF-PC LTP is not known. Immunoelectron microscopy demonstrated that βARs are expressed in PF boutons. Moreover, activation of these receptors through their agonist isoproterenol potentiated synaptic transmission in cerebellar slices from mice of either sex, an effect that was insensitive to the PKA inhibitors (H-89, KT270) but that was blocked by the Epac inhibitor ESI 05. Interestingly, prior activation of these βARs occluded PF-PC LTP, while the β1AR antagonist metoprolol blocked PF-PC LTP, which was also absent in Epac2 -/- mice. PF-PC LTP is associated with an increase in the size of the readily releasable pool (RRP) of synaptic vesicles, consistent with the isoproterenol-induced increase in vesicle docking in cerebellar slices. Thus, the βAR-mediated modulation of the release machinery and the subsequent increase in the size of the RRP contributes to PF-PC LTP.SIGNIFICANCE STATEMENT G-protein-coupled receptors modulate the release machinery, causing long-lasting changes in synaptic transmission that influence synaptic plasticity. Nevertheless, the mechanisms underlying synaptic responses to β-adrenergic receptor (βAR) activation remain poorly understood. An increase in the number of synaptic vesicles primed for exocytosis accounts for the potentiation of neurotransmitter release driven by βARs. This effect is not mediated by the canonical protein kinase A pathway but rather, through direct activation of the guanine nucleotide exchange protein Epac by cAMP. Interestingly, this βAR signaling via Epac is involved in long term potentiation at cerebellar granule cell-to-Purkinje cell synapses. Thus, the pharmacological activation of βARs modulates synaptic plasticity and opens therapeutic opportunities to control this phenomenon.
Collapse
|
30
|
Quantitative Synaptic Biology: A Perspective on Techniques, Numbers and Expectations. Int J Mol Sci 2020; 21:ijms21197298. [PMID: 33023247 PMCID: PMC7582872 DOI: 10.3390/ijms21197298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/31/2022] Open
Abstract
Synapses play a central role for the processing of information in the brain and have been analyzed in countless biochemical, electrophysiological, imaging, and computational studies. The functionality and plasticity of synapses are nevertheless still difficult to predict, and conflicting hypotheses have been proposed for many synaptic processes. In this review, we argue that the cause of these problems is a lack of understanding of the spatiotemporal dynamics of key synaptic components. Fortunately, a number of emerging imaging approaches, going beyond super-resolution, should be able to provide required protein positions in space at different points in time. Mathematical models can then integrate the resulting information to allow the prediction of the spatiotemporal dynamics. We argue that these models, to deal with the complexity of synaptic processes, need to be designed in a sufficiently abstract way. Taken together, we suggest that a well-designed combination of imaging and modelling approaches will result in a far more complete understanding of synaptic function than currently possible.
Collapse
|
31
|
Ginger L, Malsam J, Sonnen AFP, Morado D, Scheutzow A, Söllner TH, Briggs JAG. Arrangements of proteins at reconstituted synaptic vesicle fusion sites depend on membrane separation. FEBS Lett 2020; 594:3450-3463. [PMID: 32860428 PMCID: PMC7711843 DOI: 10.1002/1873-3468.13916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/16/2020] [Accepted: 08/09/2020] [Indexed: 11/18/2022]
Abstract
Synaptic vesicle proteins, including N‐ethylmaleimide‐sensitive factor attachment protein receptors (SNAREs), Synaptotagmin‐1 and Complexin, are responsible for controlling the synchronised fusion of synaptic vesicles with the presynaptic plasma membrane in response to elevated cytosolic calcium levels. A range of structures of SNAREs and their regulatory proteins have been elucidated, but the exact organisation of these proteins at synaptic junction membranes remains elusive. Here, we have used cryoelectron tomography to investigate the arrangement of synaptic proteins in an in vitro reconstituted fusion system. We found that the separation between vesicle and target membranes strongly correlates with the organisation of protein complexes at junctions. At larger membrane separations, protein complexes assume a ‘clustered’ distribution at the docking site, inducing a protrusion in the target membrane. As the membrane separation decreases, protein complexes become displaced radially outwards and assume a ‘ring‐like’ arrangement. Our findings indicate that docked vesicles can possess a wide range of protein complex numbers and be heterogeneous in their protein arrangements.
Collapse
Affiliation(s)
- Lucy Ginger
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Joerg Malsam
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Andreas F-P Sonnen
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | | | | - John A G Briggs
- MRC Laboratory of Molecular Biology, Cambridge, UK.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
32
|
Nosov G, Kahms M, Klingauf J. The Decade of Super-Resolution Microscopy of the Presynapse. Front Synaptic Neurosci 2020; 12:32. [PMID: 32848695 PMCID: PMC7433402 DOI: 10.3389/fnsyn.2020.00032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 07/21/2020] [Indexed: 01/05/2023] Open
Abstract
The presynaptic compartment of the chemical synapse is a small, yet extremely complex structure. Considering its size, most methods of optical microscopy are not able to resolve its nanoarchitecture and dynamics. Thus, its ultrastructure could only be studied by electron microscopy. In the last decade, new methods of optical superresolution microscopy have emerged allowing the study of cellular structures and processes at the nanometer scale. While this is a welcome addition to the experimental arsenal, it has necessitated careful analysis and interpretation to ensure the data obtained remains artifact-free. In this article we review the application of nanoscopic techniques to the study of the synapse and the progress made over the last decade with a particular focus on the presynapse. We find to our surprise that progress has been limited, calling for imaging techniques and probes that allow dense labeling, multiplexing, longer imaging times, higher temporal resolution, while at least maintaining the spatial resolution achieved thus far.
Collapse
Affiliation(s)
- Georgii Nosov
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany.,CIM-IMPRS Graduate Program in Münster, Münster, Germany
| | - Martin Kahms
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Jurgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| |
Collapse
|
33
|
Siddig S, Aufmkolk S, Doose S, Jobin ML, Werner C, Sauer M, Calebiro D. Super-resolution imaging reveals the nanoscale organization of metabotropic glutamate receptors at presynaptic active zones. SCIENCE ADVANCES 2020; 6:eaay7193. [PMID: 32494600 PMCID: PMC7159906 DOI: 10.1126/sciadv.aay7193] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/22/2020] [Indexed: 05/12/2023]
Abstract
G protein-coupled receptors (GPCRs) play a fundamental role in the modulation of synaptic transmission. A pivotal example is provided by the metabotropic glutamate receptor type 4 (mGluR4), which inhibits glutamate release at presynaptic active zones (AZs). However, how GPCRs are organized within AZs to regulate neurotransmission remains largely unknown. Here, we applied two-color super-resolution imaging by direct stochastic optical reconstruction microscopy (dSTORM) to investigate the nanoscale organization of mGluR4 at parallel fiber AZs in the mouse cerebellum. We find an inhomogeneous distribution, with multiple nanodomains inside AZs, each containing, on average, one to two mGluR4 subunits. Within these nanodomains, mGluR4s are often localized in close proximity to voltage-dependent CaV2.1 channels and Munc-18-1, which are both essential for neurotransmitter release. These findings provide previously unknown insights into the molecular organization of GPCRs at AZs, suggesting a likely implication of a close association between mGluR4 and the secretory machinery in modulating synaptic transmission.
Collapse
Affiliation(s)
- Sana Siddig
- Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
- Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Sarah Aufmkolk
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
- Department of Neurology & Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sören Doose
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Marie-Lise Jobin
- Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
| | - Christian Werner
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
- Corresponding author. (M.S.); (D.C.)
| | - Davide Calebiro
- Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, UK
- Corresponding author. (M.S.); (D.C.)
| |
Collapse
|
34
|
Bowers MR, Reist NE. Synaptotagmin: Mechanisms of an electrostatic switch. Neurosci Lett 2020; 722:134834. [DOI: 10.1016/j.neulet.2020.134834] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 02/09/2023]
|
35
|
Template-free detection and classification of membrane-bound complexes in cryo-electron tomograms. Nat Methods 2020; 17:209-216. [PMID: 31907446 DOI: 10.1038/s41592-019-0675-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/11/2019] [Indexed: 01/12/2023]
Abstract
With faithful sample preservation and direct imaging of fully hydrated biological material, cryo-electron tomography provides an accurate representation of molecular architecture of cells. However, detection and precise localization of macromolecular complexes within cellular environments is aggravated by the presence of many molecular species and molecular crowding. We developed a template-free image processing procedure for accurate tracing of complex networks of densities in cryo-electron tomograms, a comprehensive and automated detection of heterogeneous membrane-bound complexes and an unsupervised classification (PySeg). Applications to intact cells and isolated endoplasmic reticulum (ER) allowed us to detect and classify small protein complexes. This classification provided sufficiently homogeneous particle sets and initial references to allow subsequent de novo subtomogram averaging. Spatial distribution analysis showed that ER complexes have different localization patterns forming nanodomains. Therefore, this procedure allows a comprehensive detection and structural analysis of complexes in situ.
Collapse
|
36
|
Ranjbari E, Majdi S, Ewing A. Analytical Techniques: Shedding Light upon Nanometer-Sized Secretory Vesicles. TRENDS IN CHEMISTRY 2019. [DOI: 10.1016/j.trechm.2019.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
37
|
Critical Analysis of Particle Detection Artifacts in Synaptosome Flow Cytometry. eNeuro 2019; 6:ENEURO.0009-19.2019. [PMID: 31118205 PMCID: PMC6565374 DOI: 10.1523/eneuro.0009-19.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/01/2019] [Accepted: 04/27/2019] [Indexed: 11/21/2022] Open
Abstract
Flow cytometry and fluorescence-activated sorting are powerful techniques that hold great promise for studying heterogeneous populations of submicron particles such as synaptosomes, but many technical challenges arise in these experiments. To date, most flow cytometry studies of synaptosomes have relied on particle detection using forward scatter (FSC) measurements and size estimation with polystyrene (PS) bead standards. However, these practices have serious limitations, and special care must be taken to overcome the poor sensitivity of conventional flow cytometers in the analysis of submicron particles. Technical artifacts can confound these experiments, especially the detection of multiple particles as a single event. Here, we compared analysis of P2 crude synaptosomal preparations from murine forebrain on multiple flow cytometers using both FSC-triggered and fluorescence-triggered detection. We implemented multicolor fluorescent dye-based assays to quantify coincident particle detection and aggregation, and we assessed the false colocalization of antigens in immunostaining analyses. Our results demonstrate that fluorescence triggering and proper dilution can control for coincident particle detection, but not particle aggregation. We confirmed previous studies showing that FSC-based size estimation with PS beads underestimates biological particle size, and we identified pervasive aggregation in the FSC range analyzed in most synaptosome flow cytometry studies. We found that analyzing P2 samples in sucrose/EDTA/tris (SET) buffer reduces aggregation compared to PBS, but does not completely eliminate the presence of aggregates, especially in immunostaining experiments. Our study highlights challenges and pitfalls in synaptosome flow cytometry and provides a methodological framework for future studies.
Collapse
|
38
|
Jung JH. Synaptic Vesicles Having Large Contact Areas with the Presynaptic Membrane are Preferentially Hemifused at Active Zones of Frog Neuromuscular Junctions Fixed during Synaptic Activity. Int J Mol Sci 2019; 20:ijms20112692. [PMID: 31159267 PMCID: PMC6600287 DOI: 10.3390/ijms20112692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/23/2019] [Accepted: 05/29/2019] [Indexed: 12/29/2022] Open
Abstract
Synaptic vesicles dock on the presynaptic plasma membrane of axon terminals and become ready to fuse with the presynaptic membrane or primed. Fusion of the vesicle membrane and presynaptic membrane results in the formation of a pore between the membranes, through which the vesicle’s neurotransmitter is released into the synaptic cleft. A recent electron tomography study on frog neuromuscular junctions fixed at rest showed that there is no discernible gap between or merging of the membrane of docked synaptic vesicles with the presynaptic membrane, however, the extent of the contact area between the membrane of docked synaptic vesicles and the presynaptic membrane varies 10-fold with a normal distribution. The study also showed that when the neuromuscular junctions are fixed during repetitive electrical nerve stimulation, the portion of large contact areas in the distribution is reduced compared to the portion of small contact areas, suggesting that docked synaptic vesicles with the largest contact areas are greatly primed to fuse with the membrane. Furthermore, the finding of several hemifused synaptic vesicles among the docked vesicles was briefly reported. Here, the spatial relationship of 81 synaptic vesicles with the presynaptic membrane at active zones of the neuromuscular junctions fixed during stimulation is described in detail. For the most of the vesicles, the combined thickness of each of their contact sites was not different from the sum of the membrane thicknesses of the vesicle membrane and presynaptic membrane, similar to the docked vesicles at active zones of the resting neuromuscular junctions. However, the combined membrane thickness of a small portion of the vesicles was considerably less than the sum of the membrane thicknesses, indicating that the membranes at their contact sites were fixed in a state of hemifusion. Moreover, the hemifused vesicles were found to have large contact areas with the presynaptic membrane. These findings support the recently proposed hypothesis that, at frog neuromuscular junctions, docked synaptic vesicles with the largest contact areas are most primed for fusion with the presynaptic membrane, and that hemifusion is a fusion intermediate step of the vesicle membrane with the presynaptic membrane for synaptic transmission.
Collapse
Affiliation(s)
- Jae Hoon Jung
- Department of Biology, Texas A&M University, College Station, TX 77845, USA.
- Department of Physics, Stanford University School of Humanities and Sciences, Stanford, CA 94309, USA.
| |
Collapse
|
39
|
Chakrabarti R, Wichmann C. Nanomachinery Organizing Release at Neuronal and Ribbon Synapses. Int J Mol Sci 2019; 20:E2147. [PMID: 31052288 PMCID: PMC6539712 DOI: 10.3390/ijms20092147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
A critical aim in neuroscience is to obtain a comprehensive view of how regulated neurotransmission is achieved. Our current understanding of synapses relies mainly on data from electrophysiological recordings, imaging, and molecular biology. Based on these methodologies, proteins involved in a synaptic vesicle (SV) formation, mobility, and fusion at the active zone (AZ) membrane have been identified. In the last decade, electron tomography (ET) combined with a rapid freezing immobilization of neuronal samples opened a window for understanding the structural machinery with the highest spatial resolution in situ. ET provides significant insights into the molecular architecture of the AZ and the organelles within the presynaptic nerve terminal. The specialized sensory ribbon synapses exhibit a distinct architecture from neuronal synapses due to the presence of the electron-dense synaptic ribbon. However, both synapse types share the filamentous structures, also commonly termed as tethers that are proposed to contribute to different steps of SV recruitment and exocytosis. In this review, we discuss the emerging views on the role of filamentous structures in SV exocytosis gained from ultrastructural studies of excitatory, mainly central neuronal compared to ribbon-type synapses with a focus on inner hair cell (IHC) ribbon synapses. Moreover, we will speculate on the molecular entities that may be involved in filament formation and hence play a crucial role in the SV cycle.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
- Collaborative Research Center 1286 "Quantitative Synaptology", 37099 Göttingen, Germany.
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
40
|
Brunger AT, Choi UB, Lai Y, Leitz J, White KI, Zhou Q. The pre-synaptic fusion machinery. Curr Opin Struct Biol 2019; 54:179-188. [PMID: 30986753 PMCID: PMC6939388 DOI: 10.1016/j.sbi.2019.03.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 11/26/2022]
Abstract
Here, we review recent insights into the neuronal presynaptic fusion machinery that releases neurotransmitter molecules into the synaptic cleft upon stimulation. The structure of the pre-fusion state of the SNARE/complexin-1/synaptotagmin-1 synaptic protein complex suggests a new model for the initiation of fast Ca2+-triggered membrane fusion. Functional studies have revealed roles of the essential factors Munc18 and Munc13, demonstrating that a part of their function involves the proper assembly of synaptic protein complexes. Near-atomic resolution structures of the NSF/αSNAP/SNARE complex provide first glimpses of the molecular machinery that disassembles the SNARE complex during the synaptic vesicle cycle. These structures show how this machinery captures the SNARE substrate and provide clues as to a possible processing mechanism.
Collapse
Affiliation(s)
- Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA.
| | - Ucheor B Choi
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Ying Lai
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Kristopher Ian White
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Qiangjun Zhou
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| |
Collapse
|
41
|
Zuber B, Lučić V. Molecular architecture of the presynaptic terminal. Curr Opin Struct Biol 2019; 54:129-138. [PMID: 30925443 DOI: 10.1016/j.sbi.2019.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/26/2019] [Indexed: 10/27/2022]
Abstract
Neurotransmitter release at the presynaptic terminal is one of the fundamental processes in neuronal communication. It is a complex process comprising signaling pathways that exert a precise spatio-temporal coordination to prepare and bring synaptic vesicles to exocytosis. While many molecular components involved have been identified, their direct observation at different stages of the neurotransmitter release is lacking. Three-dimensional imaging by electron tomography provided remarkable views of the synaptic vesicles and the cytomatrix. Imaging fully hydrated, vitrified samples allowed a direct visualization, precise localization and a quantitative characterization of pleomorphic synaptic vesicle-bound complexes in situ, as well as the elucidation of their function in the neurotransmitter release.
Collapse
Affiliation(s)
- Benoît Zuber
- Institute of Anatomy, University of Bern, Bern 3012, Switzerland
| | - Vladan Lučić
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
42
|
Scholz N, Ehmann N, Sachidanandan D, Imig C, Cooper BH, Jahn O, Reim K, Brose N, Meyer J, Lamberty M, Altrichter S, Bormann A, Hallermann S, Pauli M, Heckmann M, Stigloher C, Langenhan T, Kittel RJ. Complexin cooperates with Bruchpilot to tether synaptic vesicles to the active zone cytomatrix. J Cell Biol 2019; 218:1011-1026. [PMID: 30782781 PMCID: PMC6400551 DOI: 10.1083/jcb.201806155] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 12/14/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
By performing an in vivo screen in Drosophila melanogaster, Scholz, Ehmann, et al. identify Complexin as a functional interaction partner of Bruchpilot. The two proteins mediate a physical attachment of synaptic vesicles to the active zone cytomatrix and promote rapid, sustained synaptic transmission. Information processing by the nervous system depends on neurotransmitter release from synaptic vesicles (SVs) at the presynaptic active zone. Molecular components of the cytomatrix at the active zone (CAZ) regulate the final stages of the SV cycle preceding exocytosis and thereby shape the efficacy and plasticity of synaptic transmission. Part of this regulation is reflected by a physical association of SVs with filamentous CAZ structures via largely unknown protein interactions. The very C-terminal region of Bruchpilot (Brp), a key component of the Drosophila melanogaster CAZ, participates in SV tethering. Here, we identify the conserved SNARE regulator Complexin (Cpx) in an in vivo screen for molecules that link the Brp C terminus to SVs. Brp and Cpx interact genetically and functionally. Both proteins promote SV recruitment to the Drosophila CAZ and counteract short-term synaptic depression. Analyzing SV tethering to active zone ribbons of cpx3 knockout mice supports an evolutionarily conserved role of Cpx upstream of SNARE complex assembly.
Collapse
Affiliation(s)
- Nicole Scholz
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany.,Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Leipzig University, Leipzig, Germany
| | - Nadine Ehmann
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany.,Department of Animal Physiology, Institute of Biology, Leipzig University, Leipzig, Germany.,Carl Ludwig Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Divya Sachidanandan
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kerstin Reim
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Jutta Meyer
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Germany
| | - Marius Lamberty
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany.,Department of Animal Physiology, Institute of Biology, Leipzig University, Leipzig, Germany.,Carl Ludwig Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Steffen Altrichter
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany.,Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Leipzig University, Leipzig, Germany
| | - Anne Bormann
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Leipzig University, Leipzig, Germany
| | - Stefan Hallermann
- Carl Ludwig Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Martin Pauli
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany
| | - Manfred Heckmann
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany
| | | | - Tobias Langenhan
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany .,Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Leipzig University, Leipzig, Germany
| | - Robert J Kittel
- Institute of Physiology, Department of Neurophysiology, University of Würzburg, Würzburg, Germany .,Department of Animal Physiology, Institute of Biology, Leipzig University, Leipzig, Germany.,Carl Ludwig Institute for Physiology, Leipzig University, Leipzig, Germany
| |
Collapse
|
43
|
|
44
|
Kroll J, Özçete ÖD, Jung S, Maritzen T, Milosevic I, Wichmann C, Moser T. AP180 promotes release site clearance and clathrin-dependent vesicle reformation in mouse cochlear inner hair cells. J Cell Sci 2019; 133:jcs.236737. [DOI: 10.1242/jcs.236737] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/09/2019] [Indexed: 12/18/2022] Open
Abstract
High-throughput neurotransmission at ribbon synapses of cochlear inner hair cells (IHCs) requires tight coupling of neurotransmitter release and balanced recycling of synaptic vesicles (SVs) as well as rapid restoration of release sites. Here, we examined the role of the adaptor protein AP180 for IHC synaptic transmission in AP180-KO mice using high-pressure freezing and electron tomography, confocal microscopy, patch-clamp membrane-capacitance measurements and systems physiology. AP180 was found predominantly at the synaptic pole of IHCs. AP180-deficient IHCs had severely reduced SV numbers, slowed endocytic membrane retrieval, and accumulated endocytic intermediates near ribbon synapses, indicating that AP180 is required for clathrin-dependent endocytosis and SV reformation in IHCs. Moreover, AP180 deletion led to a high prevalence of SVs in a multi-tethered or docked state after stimulation, a reduced rate of SV replenishment, and a hearing impairment. We conclude that, in addition to its role in clathrin recruitment, AP180 contributes to release site clearance in IHCs.
Collapse
Affiliation(s)
- Jana Kroll
- Synaptic Vesicle Dynamics Group, European Neuroscience Institute Göttingen – A Joint Initiative of the University Medical Center Göttingen and the Max-Planck-Society, Göttingen, Germany
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Özge Demet Özçete
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sangyong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Neuro Modulation and Neuro Circuitry Group, Singapore Bioimaging Consortium (SBIC), Biomedical Sciences Institutes, 138667 Singapore
| | - Tanja Maritzen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Ira Milosevic
- Synaptic Vesicle Dynamics Group, European Neuroscience Institute Göttingen – A Joint Initiative of the University Medical Center Göttingen and the Max-Planck-Society, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Moser
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
45
|
Pangrsic T, Vogl C. Balancing presynaptic release and endocytic membrane retrieval at hair cell ribbon synapses. FEBS Lett 2018; 592:3633-3650. [PMID: 30251250 DOI: 10.1002/1873-3468.13258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 11/07/2022]
Abstract
The timely and reliable processing of auditory and vestibular information within the inner ear requires highly sophisticated sensory transduction pathways. On a cellular level, these demands are met by hair cells, which respond to sound waves - or alterations in body positioning - by releasing glutamate-filled synaptic vesicles (SVs) from their presynaptic active zones with unprecedented speed and exquisite temporal fidelity, thereby initiating the auditory and vestibular pathways. In order to achieve this, hair cells have developed anatomical and molecular specializations, such as the characteristic and name-giving 'synaptic ribbons' - presynaptically anchored dense bodies that tether SVs prior to release - as well as other unique or unconventional synaptic proteins. The tightly orchestrated interplay between these molecular components enables not only ultrafast exocytosis, but similarly rapid and efficient compensatory endocytosis. So far, the knowledge of how endocytosis operates at hair cell ribbon synapses is limited. In this Review, we summarize recent advances in our understanding of the SV cycle and molecular anatomy of hair cell ribbon synapses, with a focus on cochlear inner hair cells.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, University Medical Center Göttingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, University Medical Center Göttingen, Germany
| |
Collapse
|
46
|
Jung JH, Szule JA, Stouder K, Marshall RM, McMahan UJ. Active Zone Material-Directed Orientation, Docking, and Fusion of Dense Core Vesicles Alongside Synaptic Vesicles at Neuromuscular Junctions. Front Neuroanat 2018; 12:72. [PMID: 30271328 PMCID: PMC6146030 DOI: 10.3389/fnana.2018.00072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
Active zone material is an organelle that is common to active zones along the presynaptic membrane of chemical synapses. Electron tomography on active zones at frog neuromuscular junctions has provided evidence that active zone material directs the docking of synaptic vesicles (SVs) on the presynaptic membrane at this synapse. Certain active zone material macromolecules connect to stereotypically arranged macromolecules in the membrane of undocked SVs, stably orienting a predetermined fusion domain of the vesicle membrane toward the presynaptic membrane while bringing and holding the two membranes together. Docking of the vesicles is required for the impulse-triggered vesicle membrane-presynaptic membrane fusion that releases the vesicles’ neurotransmitter into the synaptic cleft. As at other synapses, axon terminals at frog neuromuscular junctions contain, in addition to SVs, vesicles that are larger, are much less frequent and, when viewed by electron microscopy, have a distinctive electron dense core. Dense core vesicles at neuromuscular junctions are likely to contain peptides that are released into the synaptic cleft to regulate formation, maintenance and behavior of cellular apparatus essential for synaptic impulse transmission. We show by electron tomography on axon terminals of frog neuromuscular junctions fixed at rest and during repetitive impulse transmission that dense core vesicles selectively dock on and fuse with the presynaptic membrane alongside SVs at active zones, and that active zone material connects to the dense core vesicles undergoing these processes in the same way it connects to SVs. We conclude that undocked dense core vesicles have a predetermined fusion domain, as do undocked SVs, and that active zone material directs oriented docking and fusion of these different vesicle types at active zones of the presynaptic membrane by similar macromolecular interactions.
Collapse
Affiliation(s)
- Jae H Jung
- Department of Biology, Texas A&M University, College Station, TX, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford University, Stanford, CA, United States
| | - Joseph A Szule
- Department of Biology, Texas A&M University, College Station, TX, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford University, Stanford, CA, United States
| | - Kylee Stouder
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Robert M Marshall
- Department of Biology, Texas A&M University, College Station, TX, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford University, Stanford, CA, United States
| | - Uel J McMahan
- Department of Biology, Texas A&M University, College Station, TX, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
47
|
Chakrabarti R, Michanski S, Wichmann C. Vesicle sub-pool organization at inner hair cell ribbon synapses. EMBO Rep 2018; 19:embr.201744937. [PMID: 30201800 DOI: 10.15252/embr.201744937] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/07/2018] [Accepted: 08/21/2018] [Indexed: 02/02/2023] Open
Abstract
The afferent inner hair cell synapse harbors the synaptic ribbon, which ensures a constant vesicle supply. Synaptic vesicles (SVs) are arranged in morphologically discernable pools, linked via filaments to the ribbon or the presynaptic membrane. We propose that filaments play a major role in SV resupply and exocytosis at the ribbon. Using advanced electron microscopy, we demonstrate that SVs are organized in sub-pools defined by the filament number per vesicle and its connections. Upon stimulation, SVs increasingly linked to other vesicles and to the ribbon, whereas single-tethered SVs dominated at the membrane. Mutant mice for the hair cell protein otoferlin (pachanga, Otof Pga/Pga ) are profoundly deaf with reduced sustained release, serving as a model to investigate the SV replenishment at IHCs. Upon stimulation, multiple-tethered and docked vesicles (rarely observed in wild-type) accumulated at Otof Pga/Pga active zones due to an impairment downstream of docking. Conclusively, vesicles are organized in sub-pools at ribbon-type active zones by filaments to support vesicle supply, transport, and finally release.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany.,Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany.,Georg-August University School of Science, Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany .,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany
| |
Collapse
|
48
|
Neumüller J. Electron tomography-a tool for ultrastructural 3D visualization in cell biology and histology. Wien Med Wochenschr 2018; 168:322-329. [PMID: 30084092 PMCID: PMC6132546 DOI: 10.1007/s10354-018-0646-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Electron tomography (ET) was developed to overcome some of the problems associated reconstructing three-dimensional (3D) images from 2D election microscopy data from ultrathin slices. Virtual sections of semithin sample are obtained by incremental rotation of the target and this information is used to assemble a 3D image. Herein, we provide an instruction to ET including the physical principle, possibilities, and limitations. We review the development of innovative methods and highlight important investigations performed in our department and with our collaborators. ET has opened up the third dimension at the ultrastructural level and represents a milestone in structural molecular biology.
Collapse
Affiliation(s)
- Josef Neumüller
- Center of Anatomy and Cell Biology, Department of Cell and Developmental Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria.
| |
Collapse
|
49
|
Brunger AT, Leitz J, Zhou Q, Choi UB, Lai Y. Ca 2+-Triggered Synaptic Vesicle Fusion Initiated by Release of Inhibition. Trends Cell Biol 2018; 28:631-645. [PMID: 29706534 PMCID: PMC6056330 DOI: 10.1016/j.tcb.2018.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/17/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022]
Abstract
Recent structural and functional studies of the synaptic vesicle fusion machinery suggest an inhibited tripartite complex consisting of neuronal soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs), synaptotagmin, and complexin prior to Ca2+-triggered synaptic vesicle fusion. We speculate that Ca2+-triggered fusion commences with the release of inhibition by Ca2+ binding to synaptotagmin C2 domains. Subsequently, fusion is assisted by SNARE complex zippering and by active membrane remodeling properties of synaptotagmin. This additional, inhibitory role of synaptotagmin may be a general principle since other recent studies suggest that Ca2+ binding to extended synaptotagmin C2 domains enables lipid transport by releasing an inhibited state of the system, and that Munc13 may nominally be in an inhibited state, which is released upon Ca2+ binding to one of its C2 domains.
Collapse
Affiliation(s)
- Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Qiangjun Zhou
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Ucheor B Choi
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Ying Lai
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
50
|
Schrod N, Vanhecke D, Laugks U, Stein V, Fukuda Y, Schaffer M, Baumeister W, Lucic V. Pleomorphic linkers as ubiquitous structural organizers of vesicles in axons. PLoS One 2018; 13:e0197886. [PMID: 29864134 PMCID: PMC5986143 DOI: 10.1371/journal.pone.0197886] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/10/2018] [Indexed: 11/30/2022] Open
Abstract
Many cellular processes depend on a precise structural organization of molecular components. Here, we established that neurons grown in culture provide a suitable system for in situ structural investigations of cellular structures by cryo-electron tomography, a method that allows high resolution, three-dimensional imaging of fully hydrated, vitrified cellular samples. A higher level of detail of cellular components present in our images allowed us to quantitatively characterize presynaptic and cytoskeletal organization, as well as structures involved in axonal transport and endocytosis. In this way we provide a structural framework into which information from other methods need to fit. Importantly, we show that short pleomorphic linkers (tethers and connectors) extensively interconnect different types of spherical vesicles and other lipid membranes in neurons imaged in a close-to-native state. These linkers likely serve to organize and precisely position vesicles involved in endocytosis, axonal transport and synaptic release. Hence, structural interactions via short linkers may serve as ubiquitous vesicle organizers in neuronal cells.
Collapse
Affiliation(s)
- Nikolas Schrod
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Dimitri Vanhecke
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Ulrike Laugks
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Valentin Stein
- Institute of Physiology II, University of Bonn, Bonn, Germany
| | - Yoshiyuki Fukuda
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Miroslava Schaffer
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Wolfgang Baumeister
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| | - Vladan Lucic
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Germany
| |
Collapse
|