1
|
Sun X, Zhang Y, Wang H, Pu X, Yuan X, Liang Y, Liu H, Wang X, Lu H. N 6-methyladenosine modification of RIMS binding protein 2 promotes head and neck squamous cell carcinoma proliferation and radiotherapy tolerance through endoplasmic reticulum stress. Cancer Gene Ther 2025; 32:122-135. [PMID: 39653741 DOI: 10.1038/s41417-024-00863-8] [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: 08/11/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/29/2025]
Abstract
Insulin-like growth factor binding protein 2 (IGF2BP2) fulfills a key role in the development of head and neck squamous cell carcinoma (HNSCC). Radiotherapy is an effective method to treat HNSCC; however, radiation resistance is the main reason for treatment failure. At present, the carcinogenic role of IGF2BP2 in terms of the proliferation of HNSCC and the radioresistance of its therapy remain poorly understood. In the present study, patients with HNSCC with higher IGF2BP2 expression levels were associated with shorter survival times. IGF2BP2 is significantly upregulated in HNSCC cells compared with irradiated cell. Based on functional studies, IGF2BP2 was found to promote HNSCC cell proliferation and tolerance to radiotherapy both in vitro and in vivo. In terms of the underlying mechanism, RIMS binding protein 2 (RIMBP2) was found to be highly expressed in HNSCC and to promote the proliferation of HNSCC and radiotherapy resistance. RIMBP2 was shown to be a direct target of IGF2BP2, activating endoplasmic reticulum stress in HNSCC. In addition, it has been demonstrated that IGF2BP2, as m6A reader, is able to promote RIMBP2 stability via binding to m6A sites in the RIMBP2-coding sequence region. Therefore, the present study has unveiled a potential mechanism via which IGF2BP2 promotes HNSCC development and radiotherapy resistance; moreover, from a therapeutic perspective, IGF2BP2 may serve as a potential therapeutic target and a valuable prognostic biomarker for patients with HNSCC who have developed tolerance towards radiotherapy.
Collapse
Affiliation(s)
- Xinyu Sun
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Yanshu Zhang
- Department of Otorhinolaryngology, Yancheng Clinical Medical College of Jiangsu University/The First people's Hospital of Yancheng, 224001, Yancheng, Jiangsu Province, China
| | - Huirong Wang
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Xi Pu
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Xiao Yuan
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Yuntong Liang
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Hao Liu
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Xu Wang
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China
| | - Hanqiang Lu
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, Jiangsu Province, China.
| |
Collapse
|
2
|
Maglione M, Sigrist SJ. Imaging Synapse Ultrastructure and Organization with STED Microscopy. Methods Mol Biol 2025; 2910:135-144. [PMID: 40220098 DOI: 10.1007/978-1-0716-4446-1_9] [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] [Indexed: 04/14/2025]
Abstract
Determining the localization of proteins within a cell and their possible interactions is highly relevant to understand their functionality. Nevertheless, subcellular structures of interest in neurobiology, most importantly synapses with their pre- and postsynaptic compartments, are usually smaller than the resolution limit of conventional light microscopy. Indeed, diffraction of light limits to roughly half of the wavelength of light the resolution of a conventional light microscope. In this regard, super-resolution light microscopy (SRLM) techniques have emerged, achieving even more than ten times the resolution of conventional light microscopy, thus allowing to resolve subsynaptic structures also in situ. Importantly, stimulated emission depletion (STED) microscopy has been extensively used to image in situ the nanoscale organization of presynaptic compartments, such as the area of the presynaptic plasma membrane where synaptic vesicles fuse to release neurotransmitters, the so-called active zone. In this article, we outline a method to determine the localization of active zone scaffolding key players relative to voltage-gated calcium channels within the presynaptic active zone by time-gated STED (gSTED) microscopy in situ.
Collapse
Affiliation(s)
- Marta Maglione
- Institute for Biology, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
- Institute for Chemistry and Biochemistry, SupraFAB, Freie Universität Berlin, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology, Freie Universität Berlin, Berlin, Germany.
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
3
|
Marneffe C, Moreira-de-Sá A, Lecomte S, Erhardt A, Mulle C. Short term plasticity at hippocampal mossy fiber synapses. Neuroscience 2024:S0306-4522(24)00497-4. [PMID: 39332701 DOI: 10.1016/j.neuroscience.2024.09.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Short-term synaptic plasticity refers to the regulation of synapses by their past activity on time scales of milliseconds to minutes. Hippocampal mossy fiber synapses onto CA3 pyramidal cells (Mf-CA3 synapses) are endowed with remarkable forms of short-term synaptic plasticity expressed as facilitation of synaptic release by a factor of up to ten-fold. Three main forms of short-term plasticity are distinguished: 1) Frequency facilitation, which includes low frequency facilitation and train facilitation, operating in the range of tens of milliseconds to several seconds; 2) Post-tetanic potentiation triggered by trains of high frequency stimulation, which lasts several minutes; 3) Finally, depolarization-induced potentiation of excitation, based on retrograde signaling, with an onset and offset of several minutes. Here we describe the proposed mechanisms for short-term plasticity, mainly based on the kinetics of presynaptic Ca2+ transients and the Ca2+ sensor synaptotagmin 7, on cAMP-dependent mechanisms and readily releasable vesicle pool, and on the regulation of presynaptic K+ channels. We then review evidence for a physiological function of short-term plasticity of Mf-CA3 synapses in information transfer between the dentate gyrus and CA3 in conditions of natural spiking, and discuss how short-term plasticity counteracts robust feedforward inhibition in a frequency-dependent manner. Although DG-CA3 connections have long been proposed to play a role in memory, direct evidence for an implication of short-term plasticity at Mf-CA3 synapses is mostly lacking. The mechanistic knowledge gained on short-term plasticity at Mf-CA3 synapses should help in designing future experiments to directly test how this evolutionary conserved feature controls hippocampal circuit function in behavioural conditions.
Collapse
Affiliation(s)
- Catherine Marneffe
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Ana Moreira-de-Sá
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Simon Lecomte
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Anaël Erhardt
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, France; University of Bordeaux, F-33000 Bordeaux, France.
| |
Collapse
|
4
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Distinct active zone protein machineries mediate Ca 2+ channel clustering and vesicle priming at hippocampal synapses. Nat Neurosci 2024; 27:1680-1694. [PMID: 39160372 PMCID: PMC11682530 DOI: 10.1038/s41593-024-01720-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/28/2024] [Indexed: 08/21/2024]
Abstract
Action potentials trigger neurotransmitter release at the presynaptic active zone with spatiotemporal precision. This is supported by protein machinery that mediates synaptic vesicle priming and clustering of CaV2 Ca2+ channels nearby. One model posits that scaffolding proteins directly tether vesicles to CaV2s; however, here we find that at mouse hippocampal synapses, CaV2 clustering and vesicle priming are executed by separate machineries. CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins but distinct interaction motifs independently execute these functions. In transfected cells, Liprin-α and RIM form co-assemblies that are separate from CaV2-organizing complexes. At synapses, Liprin-α1-Liprin-α4 knockout impairs vesicle priming but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering CaV2s. We conclude that active zones consist of distinct machineries to organize CaV2s and prime vesicles, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Bär J, Fanutza T, Reimann CC, Seipold L, Grohe M, Bolter JR, Delfs F, Bucher M, Gee CE, Schweizer M, Saftig P, Mikhaylova M. Non-canonical function of ADAM10 in presynaptic plasticity. Cell Mol Life Sci 2024; 81:342. [PMID: 39123091 PMCID: PMC11335265 DOI: 10.1007/s00018-024-05327-8] [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: 02/20/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 08/12/2024]
Abstract
A Disintegrin And Metalloproteinase 10 (ADAM10) plays a pivotal role in shaping neuronal networks by orchestrating the activity of numerous membrane proteins through the shedding of their extracellular domains. Despite its significance in the brain, the specific cellular localization of ADAM10 remains not well understood due to a lack of appropriate tools. Here, using a specific ADAM10 antibody suitable for immunostainings, we observed that ADAM10 is localized to presynapses and especially enriched at presynaptic vesicles of mossy fiber (MF)-CA3 synapses in the hippocampus. These synapses undergo pronounced frequency facilitation of neurotransmitter release, a process that play critical roles in information transfer and neural computation. We demonstrate, that in conditional ADAM10 knockout mice the ability of MF synapses to undergo this type of synaptic plasticity is greatly reduced. The loss of facilitation depends on the cytosolic domain of ADAM10 and association with the calcium sensor synaptotagmin 7 rather than ADAM10's proteolytic activity. Our findings unveil a new role of ADAM10 in the regulation of synaptic vesicle exocytosis.
Collapse
Affiliation(s)
- Julia Bär
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tomas Fanutza
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Christopher C Reimann
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Lisa Seipold
- Biochemisches Institut, Christian Albrechts-Universität Kiel, 24098, Kiel, Germany
| | - Maja Grohe
- Biochemisches Institut, Christian Albrechts-Universität Kiel, 24098, Kiel, Germany
| | - Janike Rabea Bolter
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany
| | - Flemming Delfs
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Michael Bucher
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Christine E Gee
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, ZMNH, 20251, Hamburg, Germany
| | - Paul Saftig
- Biochemisches Institut, Christian Albrechts-Universität Kiel, 24098, Kiel, Germany.
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany.
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
6
|
Jánosi B, Liewald JF, Seidenthal M, Yu SC, Umbach S, Redzovic J, Rentsch D, Alcantara IC, Bergs ACF, Schneider MW, Shao J, Gottschalk A. RIM and RIM-Binding Protein Localize Synaptic CaV2 Channels to Differentially Regulate Transmission in Neuronal Circuits. J Neurosci 2024; 44:e0535222024. [PMID: 38951038 PMCID: PMC11293454 DOI: 10.1523/jneurosci.0535-22.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/16/2024] [Accepted: 05/05/2024] [Indexed: 07/03/2024] Open
Abstract
At chemical synapses, voltage-gated Ca2+ channels (VGCCs) translate electrical signals into a trigger for synaptic vesicle (SV) fusion. VGCCs and the Ca2+ microdomains they elicit must be located precisely to primed SVs to evoke rapid transmitter release. Localization is mediated by Rab3-interacting molecule (RIM) and RIM-binding proteins, which interact and bind to the C terminus of the CaV2 VGCC α-subunit. We studied this machinery at the mixed cholinergic/GABAergic neuromuscular junction of Caenorhabditis elegans hermaphrodites. rimb-1 mutants had mild synaptic defects, through loosening the anchoring of UNC-2/CaV2 and delaying the onset of SV fusion. UNC-10/RIM deletion much more severely affected transmission. Although postsynaptic depolarization was reduced, rimb-1 mutants had increased cholinergic (but reduced GABAergic) transmission, to compensate for the delayed release. This did not occur when the excitation-inhibition (E-I) balance was altered by removing GABA transmission. Further analyses of GABA defective mutants and GABAA or GABAB receptor deletions, as well as cholinergic rescue of RIMB-1, emphasized that GABA neurons may be more affected than cholinergic neurons. Thus, RIMB-1 function differentially affects excitation-inhibition balance in the different motor neurons, and RIMB-1 thus may differentially regulate transmission within circuits. Untethering the UNC-2/CaV2 channel by removing its C-terminal PDZ ligand exacerbated the rimb-1 defects, and similar phenotypes resulted from acute degradation of the CaV2 β-subunit CCB-1. Therefore, untethering of the CaV2 complex is as severe as its elimination, yet it does not abolish transmission, likely due to compensation by CaV1. Thus, robustness and flexibility of synaptic transmission emerge from VGCC regulation.
Collapse
Affiliation(s)
- Barbara Jánosi
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Jana F Liewald
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Marius Seidenthal
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Szi-Chieh Yu
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Simon Umbach
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Jasmina Redzovic
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Dennis Rentsch
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Ivan C Alcantara
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Amelie C F Bergs
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Martin W Schneider
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Jiajie Shao
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt D-60438, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute for Biophysical Chemistry, Goethe University Frankfurt, Frankfurt D-60438, Germany
| |
Collapse
|
7
|
Belghazi M, Iborra C, Toutendji O, Lasserre M, Debanne D, Goaillard JM, Marquèze-Pouey B. High-Resolution Proteomics Unravel a Native Functional Complex of Cav1.3, SK3, and Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels in Midbrain Dopaminergic Neurons. Cells 2024; 13:944. [PMID: 38891076 PMCID: PMC11172389 DOI: 10.3390/cells13110944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Pacemaking activity in substantia nigra dopaminergic neurons is generated by the coordinated activity of a variety of distinct somatodendritic voltage- and calcium-gated ion channels. We investigated whether these functional interactions could arise from a common localization in macromolecular complexes where physical proximity would allow for efficient interaction and co-regulations. For that purpose, we immunopurified six ion channel proteins involved in substantia nigra neuron autonomous firing to identify their molecular interactions. The ion channels chosen as bait were Cav1.2, Cav1.3, HCN2, HCN4, Kv4.3, and SK3 channel proteins, and the methods chosen to determine interactions were co-immunoprecipitation analyzed through immunoblot and mass spectrometry as well as proximity ligation assay. A macromolecular complex composed of Cav1.3, HCN, and SK3 channels was unraveled. In addition, novel potential interactions between SK3 channels and sclerosis tuberous complex (Tsc) proteins, inhibitors of mTOR, and between HCN4 channels and the pro-degenerative protein Sarm1 were uncovered. In order to demonstrate the presence of these molecular interactions in situ, we used proximity ligation assay (PLA) imaging on midbrain slices containing the substantia nigra, and we could ascertain the presence of these protein complexes specifically in substantia nigra dopaminergic neurons. Based on the complementary functional role of the ion channels in the macromolecular complex identified, these results suggest that such tight interactions could partly underly the robustness of pacemaking in dopaminergic neurons.
Collapse
Affiliation(s)
- Maya Belghazi
- CRN2M Centre de Recherche Neurobiologie-Neurophysiologie, CNRS, UMR7286, Aix-Marseille Université, 13015 Marseille, France;
- Institut de Microbiologie de la Méditerranée (IMM), CNRS, Aix-Marseille Université, 13009 Marseille, France
| | - Cécile Iborra
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Ophélie Toutendji
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Manon Lasserre
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Dominique Debanne
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Jean-Marc Goaillard
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
- Institut de Neurosciences de la Timone, CNRS, Aix-Marseille Université, 13005 Marseille, France
| | - Béatrice Marquèze-Pouey
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| |
Collapse
|
8
|
Davis BA, Chen HY, Ye Z, Ostlund I, Tippani M, Das D, Sripathy SR, Wang Y, Martin JM, Shim G, Panchwagh NM, Moses RL, Farinelli F, Bohlen JF, Li M, Luikart BW, Jaffe AE, Maher BJ. TCF4 Mutations Disrupt Synaptic Function Through Dysregulation of RIMBP2 in Patient-Derived Cortical Neurons. Biol Psychiatry 2024; 95:662-675. [PMID: 37573005 PMCID: PMC10858293 DOI: 10.1016/j.biopsych.2023.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Genetic variation in the TCF4 (transcription factor 4) gene is associated with risk for a variety of developmental and psychiatric conditions, which includes a syndromic form of autism spectrum disorder called Pitt-Hopkins syndrome (PTHS). TCF4 encodes an activity-dependent transcription factor that is highly expressed during cortical development and in animal models has been shown to regulate various aspects of neuronal development and function. However, our understanding of how disease-causing mutations in TCF4 confer pathophysiology in a human context is lacking. METHODS To model PTHS, we differentiated human cortical neurons from human induced pluripotent stem cells that were derived from patients with PTHS and neurotypical individuals. To identify pathophysiology and disease mechanisms, we assayed cortical neurons with whole-cell electrophysiology, Ca2+ imaging, multielectrode arrays, immunocytochemistry, and RNA sequencing. RESULTS Cortical neurons derived from patients with TCF4 mutations showed deficits in spontaneous synaptic transmission, network excitability, and homeostatic plasticity. Transcriptomic analysis indicated that these phenotypes resulted in part from altered expression of genes involved in presynaptic neurotransmission and identified the presynaptic binding protein RIMBP2 as the most differentially expressed gene in PTHS neurons. Remarkably, TCF4-dependent deficits in spontaneous synaptic transmission and network excitability were rescued by increasing RIMBP2 expression in presynaptic neurons. CONCLUSIONS Taken together, these results identify TCF4 as a critical transcriptional regulator of human synaptic development and plasticity and specifically identifies dysregulation of presynaptic function as an early pathophysiology in PTHS.
Collapse
Affiliation(s)
- Brittany A Davis
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Huei-Ying Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Zengyou Ye
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Isaac Ostlund
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Debamitra Das
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Srinidhi Rao Sripathy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Yanhong Wang
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Jacqueline M Martin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Gina Shim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Neel M Panchwagh
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Rebecca L Moses
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Federica Farinelli
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Joseph F Bohlen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Meijie Li
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland; McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Brady J Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
9
|
Vandael D, Jonas P. Structure, biophysics, and circuit function of a "giant" cortical presynaptic terminal. Science 2024; 383:eadg6757. [PMID: 38452088 DOI: 10.1126/science.adg6757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/19/2024] [Indexed: 03/09/2024]
Abstract
The hippocampal mossy fiber synapse, formed between axons of dentate gyrus granule cells and dendrites of CA3 pyramidal neurons, is a key synapse in the trisynaptic circuitry of the hippocampus. Because of its comparatively large size, this synapse is accessible to direct presynaptic recording, allowing a rigorous investigation of the biophysical mechanisms of synaptic transmission and plasticity. Furthermore, because of its placement in the very center of the hippocampal memory circuit, this synapse seems to be critically involved in several higher network functions, such as learning, memory, pattern separation, and pattern completion. Recent work based on new technologies in both nanoanatomy and nanophysiology, including presynaptic patch-clamp recording, paired recording, super-resolution light microscopy, and freeze-fracture and "flash-and-freeze" electron microscopy, has provided new insights into the structure, biophysics, and network function of this intriguing synapse. This brings us one step closer to answering a fundamental question in neuroscience: how basic synaptic properties shape higher network computations.
Collapse
Affiliation(s)
- David Vandael
- Institute of Science and Technology Austria (ISTA), A-3400 Klosterneuburg, Austria
| | - Peter Jonas
- Institute of Science and Technology Austria (ISTA), A-3400 Klosterneuburg, Austria
| |
Collapse
|
10
|
Miyano R, Sakamoto H, Hirose K, Sakaba T. RIM-BP2 regulates Ca 2+ channel abundance and neurotransmitter release at hippocampal mossy fiber terminals. eLife 2024; 12:RP90799. [PMID: 38329474 PMCID: PMC10945523 DOI: 10.7554/elife.90799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Synaptic vesicles dock and fuse at the presynaptic active zone (AZ), the specialized site for transmitter release. AZ proteins play multiple roles such as recruitment of Ca2+ channels as well as synaptic vesicle docking, priming, and fusion. However, the precise role of each AZ protein type remains unknown. In order to dissect the role of RIM-BP2 at mammalian cortical synapses having low release probability, we applied direct electrophysiological recording and super-resolution imaging to hippocampal mossy fiber terminals of RIM-BP2 knockout (KO) mice. By using direct presynaptic recording, we found the reduced Ca2+ currents. The measurements of excitatory postsynaptic currents (EPSCs) and presynaptic capacitance suggested that the initial release probability was lowered because of the reduced Ca2+ influx and impaired fusion competence in RIM-BP2 KO. Nevertheless, larger Ca2+ influx restored release partially. Consistent with presynaptic recording, STED microscopy suggested less abundance of P/Q-type Ca2+ channels at AZs deficient in RIM-BP2. Our results suggest that the RIM-BP2 regulates both Ca2+ channel abundance and transmitter release at mossy fiber synapses.
Collapse
Affiliation(s)
- Rinako Miyano
- Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| | - Hirokazu Sakamoto
- Department of Pharmacology, Graduate School of Medicine, The University of TokyoBunkyo-kuJapan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of TokyoBunkyo-kuJapan
- International Research Center for Neurointelligence (WPI-IRCN), The University of TokyoBunkyo-kuJapan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| |
Collapse
|
11
|
Pecori A, Luppieri V, Santin A, Spedicati B, Zampieri S, Cadenaro M, Girotto G, Concas MP. Clenching the Strings of Bruxism Etiopathogenesis: Association Analyses on Genetics and Environmental Risk Factors in a Deeply Characterized Italian Cohort. Biomedicines 2024; 12:304. [PMID: 38397906 PMCID: PMC10887134 DOI: 10.3390/biomedicines12020304] [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: 01/05/2024] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Bruxism is a worldwide oral health problem. Although there is a consensus about its multifactorial nature, its precise etiopathogenetic mechanisms are unclear. This study, taking advantage of a deeply characterized cohort of 769 individuals (aged 6-89 years) coming from Northern Italy's genetically isolated populations, aims to epidemiologically describe environmental risk factors for bruxism development and identify genes potentially involved through a Genome-Wide Association Study (GWAS) approach. Logistic mixed models adjusted for age and sex were performed to evaluate associations between bruxism and possible risk factors, e.g., anxiety, smoking, and alcohol and caffeine intake. A case-control GWAS (135 cases, 523 controls), adjusted for age, sex, and anxiety, was conducted to identify new candidate genes. The GTEx data analysis was performed to evaluate the identified gene expression in human body tissues. Statistical analyses determined anxiety as a bruxism risk factor (OR = 2.54; 95% CI: 1.20-5.38; p-value = 0.015), and GWAS highlighted three novel genes potentially associated with bruxism: NLGN1 (topSNP = rs2046718; p-value = 2.63 × 10-7), RIMBP2 (topSNP = rs571497947; p-value = 4.68 × 10-7), and LHFP (topSNP = rs2324342; p-value = 7.47 × 10-6). The GTEx data analysis showed their expression in brain tissues. Overall, this work provided a deeper understanding of bruxism etiopathogenesis with the long-term perspective of developing personalized therapeutic approaches for improving affected individuals' quality of life.
Collapse
Affiliation(s)
- Alessandro Pecori
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
| | - Valentina Luppieri
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Aurora Santin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Beatrice Spedicati
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Stefania Zampieri
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
| | - Milena Cadenaro
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Giorgia Girotto
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149 Trieste, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, Via dell’Istria 65, 34137 Trieste, Italy; (A.P.); (V.L.); (B.S.); (S.Z.); (M.C.); (G.G.); (M.P.C.)
| |
Collapse
|
12
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Molecular definition of distinct active zone protein machineries for Ca 2+ channel clustering and synaptic vesicle priming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564439. [PMID: 37961089 PMCID: PMC10634917 DOI: 10.1101/2023.10.27.564439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Action potentials trigger neurotransmitter release with minimal delay. Active zones mediate this temporal precision by co-organizing primed vesicles with CaV2 Ca2+ channels. The presumed model is that scaffolding proteins directly tether primed vesicles to CaV2s. We find that CaV2 clustering and vesicle priming are executed by separate machineries. At hippocampal synapses, CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins, but distinct interaction motifs independently execute these functions. In heterologous cells, Liprin-α and RIM from co-assemblies that are separate from CaV2-organizing complexes upon co-transfection. At synapses, Liprin-α1-4 knockout impairs vesicle priming, but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering of CaV2s. We conclude that active zones consist of distinct complexes to organize CaV2s and vesicle priming, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R. Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Aaron D. Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Poorna A. Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | |
Collapse
|
13
|
Eilts J, Reinhard S, Michetschläger N, Werner C, Sauer M. Enhanced synaptic protein visualization by multicolor super-resolution expansion microscopy. NEUROPHOTONICS 2023; 10:044412. [PMID: 37886043 PMCID: PMC10599331 DOI: 10.1117/1.nph.10.4.044412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/28/2023]
Abstract
Significance Understanding the organization of biomolecules into complexes and their dynamics is crucial for comprehending cellular functions and dysfunctions, particularly in neuronal networks connected by synapses. In the last two decades, various powerful super-resolution (SR) microscopy techniques have been developed that produced stunning images of synapses and their molecular organization. However, current SR microscopy methods do not permit multicolor fluorescence imaging with 20 to 30 nm spatial resolution. Aim We developed a method that enables 4-color fluorescence imaging of synaptic proteins in neurons with 20 to 30 nm lateral resolution. Approach We used post-expansion immunolabeling of eightfold expanded hippocampal neurons in combination with Airyscan and structured illumination microscopy (SIM). Results We demonstrate that post-expansion immunolabeling of approximately eightfold expanded hippocampal neurons enables efficient labeling of synaptic proteins in crowded compartments with minimal linkage error and enables in combination with Airyscan and SIM four-color three-dimensional fluorescence imaging with 20 to 30 nm lateral resolution. Using immunolabeling of Synaptobrevin 2 as an efficient marker of the vesicle pool allowed us to identify individual synaptic vesicles colocalized with Rab3-interacting molecule 1 and 2 (RIM1/2), a marker of pre-synaptic fusion sites. Conclusions Our optimized expansion microscopy approach improves the visualization and location of pre- and post-synaptic proteins and can thus provide invaluable insights into the spatial organization of proteins at synapses.
Collapse
Affiliation(s)
- Janna Eilts
- University of Würzburg, Department of Biotechnology and Biophysics, Biocenter, Würzburg, Germany
| | - Sebastian Reinhard
- University of Würzburg, Department of Biotechnology and Biophysics, Biocenter, Würzburg, Germany
| | - Nikolas Michetschläger
- University of Würzburg, Department of Biotechnology and Biophysics, Biocenter, Würzburg, Germany
| | - Christian Werner
- University of Würzburg, Department of Biotechnology and Biophysics, Biocenter, Würzburg, Germany
| | - Markus Sauer
- University of Würzburg, Department of Biotechnology and Biophysics, Biocenter, Würzburg, Germany
- University of Würzburg, Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, Würzburg, Germany
| |
Collapse
|
14
|
Izumi T. Multiple pathways and independent functional pools in insulin granule exocytosis. Genes Cells 2023; 28:471-481. [PMID: 37070774 PMCID: PMC11448364 DOI: 10.1111/gtc.13029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/19/2023]
Abstract
In contrast to synaptic vesicle exocytosis, secretory granule exocytosis follows a much longer time course, and thus allows for different prefusion states prior to stimulation. Indeed, total internal reflection fluorescence microscopy in living pancreatic β cells reveals that, prior to stimulation, either visible or invisible granules fuse in parallel during both early (first) and late (second) phases after glucose stimulation. Therefore, fusion occurs not only from granules predocked to the plasma membrane but also from those translocated from the cell interior during ongoing stimulation. Recent findings suggest that such heterogeneous exocytosis is conducted by a specific set of multiple Rab27 effectors that appear to operate on the same granule; namely, exophilin-8, granuphilin, and melanophilin play differential roles in distinct secretory pathways to final fusion. Furthermore, the exocyst, which is known to tether secretory vesicles to the plasma membrane in constitutive exocytosis, cooperatively functions with these Rab27 effectors in regulated exocytosis. In this review, the basic nature of insulin granule exocytosis will be described as a representative example of secretory granule exocytosis, followed by a discussion of the means by which different Rab27 effectors and the exocyst coordinate to regulate the entire exocytic processes in β cells.
Collapse
Affiliation(s)
- Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
15
|
Krout M, Oh KH, Xiong A, Frankel EB, Kurshan PT, Kim H, Richmond JE. C. elegans Clarinet/CLA-1 recruits RIMB-1/RIM-binding protein and UNC-13 to orchestrate presynaptic neurotransmitter release. Proc Natl Acad Sci U S A 2023; 120:e2220856120. [PMID: 37186867 PMCID: PMC10214197 DOI: 10.1073/pnas.2220856120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Synaptic transmission requires the coordinated activity of multiple synaptic proteins that are localized at the active zone (AZ). We previously identified a Caenorhabditis elegans protein named Clarinet (CLA-1) based on homology to the AZ proteins Piccolo, Rab3-interactingmolecule (RIM)/UNC-10 and Fife. At the neuromuscular junction (NMJ), cla-1 null mutants exhibit release defects that are greatly exacerbated in cla-1;unc-10 double mutants. To gain insights into the coordinated roles of CLA-1 and UNC-10, we examined the relative contributions of each to the function and organization of the AZ. Using a combination of electrophysiology, electron microscopy, and quantitative fluorescence imaging we explored the functional relationship of CLA-1 to other key AZ proteins including: RIM1, Cav2.1 channels, RIM1-binding protein, and Munc13 (C. elegans UNC-10, UNC-2, RIMB-1 and UNC-13, respectively). Our analyses show that CLA-1 acts in concert with UNC-10 to regulate UNC-2 calcium channel levels at the synapse via recruitment of RIMB-1. In addition, CLA-1 exerts a RIMB-1-independent role in the localization of the priming factor UNC-13. Thus C. elegans CLA-1/UNC-10 exhibit combinatorial effects that have overlapping design principles with other model organisms: RIM/RBP and RIM/ELKS in mouse and Fife/RIM and BRP/RBP in Drosophila. These data support a semiconserved arrangement of AZ scaffolding proteins that are necessary for the localization and activation of the fusion machinery within nanodomains for precise coupling to Ca2+ channels.
Collapse
Affiliation(s)
- Mia Krout
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
| | - Kelly H. Oh
- Department of Cell Biology and Anatomy, Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Ame Xiong
- Department of Cell Biology and Anatomy, Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Elisa B. Frankel
- Department of Genetics, Albert Einstein College of Medicine, New York, NY10461
| | - Peri T. Kurshan
- Department of Genetics, Albert Einstein College of Medicine, New York, NY10461
| | - Hongkyun Kim
- Department of Genetics, Albert Einstein College of Medicine, New York, NY10461
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
| |
Collapse
|
16
|
Zhao K, Matsunaga K, Mizuno K, Wang H, Okunishi K, Izumi T. Functional hierarchy among different Rab27 effectors involved in secretory granule exocytosis. eLife 2023; 12:82821. [PMID: 36803984 PMCID: PMC9988257 DOI: 10.7554/elife.82821] [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: 08/18/2022] [Accepted: 02/21/2023] [Indexed: 02/23/2023] Open
Abstract
The Rab27 effectors are known to play versatile roles in regulated exocytosis. In pancreatic beta cells, exophilin-8 anchors granules in the peripheral actin cortex, whereas granuphilin and melanophilin mediate granule fusion with and without stable docking to the plasma membrane, respectively. However, it is unknown whether these coexisting effectors function in parallel or in sequence to support the whole insulin secretory process. Here, we investigate their functional relationships by comparing the exocytic phenotypes in mouse beta cells simultaneously lacking two effectors with those lacking just one of them. Analyses of prefusion profiles by total internal reflection fluorescence microscopy suggest that melanophilin exclusively functions downstream of exophilin-8 to mobilize granules for fusion from the actin network to the plasma membrane after stimulation. The two effectors are physically linked via the exocyst complex. Downregulation of the exocyst component affects granule exocytosis only in the presence of exophilin-8. The exocyst and exophilin-8 also promote fusion of granules residing beneath the plasma membrane prior to stimulation, although they differentially act on freely diffusible granules and those stably docked to the plasma membrane by granuphilin, respectively. This is the first study to diagram the multiple intracellular pathways of granule exocytosis and the functional hierarchy among different Rab27 effectors within the same cell.
Collapse
Affiliation(s)
- Kunli Zhao
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Kohichi Matsunaga
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Kouichi Mizuno
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Hao Wang
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Katsuhide Okunishi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| |
Collapse
|
17
|
Ghelani T, Escher M, Thomas U, Esch K, Lützkendorf J, Depner H, Maglione M, Parutto P, Gratz S, Matkovic-Rachid T, Ryglewski S, Walter AM, Holcman D, O‘Connor Giles K, Heine M, Sigrist SJ. Interactive nanocluster compaction of the ELKS scaffold and Cacophony Ca 2+ channels drives sustained active zone potentiation. SCIENCE ADVANCES 2023; 9:eade7804. [PMID: 36800417 PMCID: PMC9937578 DOI: 10.1126/sciadv.ade7804] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/17/2023] [Indexed: 06/01/2023]
Abstract
At presynaptic active zones (AZs), conserved scaffold protein architectures control synaptic vesicle (SV) release by defining the nanoscale distribution and density of voltage-gated Ca2+ channels (VGCCs). While AZs can potentiate SV release in the minutes range, we lack an understanding of how AZ scaffold components and VGCCs engage into potentiation. We here establish dynamic, intravital single-molecule imaging of endogenously tagged proteins at Drosophila AZs undergoing presynaptic homeostatic potentiation. During potentiation, the numbers of α1 VGCC subunit Cacophony (Cac) increased per AZ, while their mobility decreased and nanoscale distribution compacted. These dynamic Cac changes depended on the interaction between Cac channel's intracellular carboxyl terminus and the membrane-close amino-terminal region of the ELKS-family protein Bruchpilot, whose distribution compacted drastically. The Cac-ELKS/Bruchpilot interaction was also needed for sustained AZ potentiation. Our single-molecule analysis illustrates how the AZ scaffold couples to VGCC nanoscale distribution and dynamics to establish a state of sustained potentiation.
Collapse
Affiliation(s)
- Tina Ghelani
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Marc Escher
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Ulrich Thomas
- Department of Cellular Neurobiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Klara Esch
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Janine Lützkendorf
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Harald Depner
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Marta Maglione
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
- Institute for Chemistry and Biochemistry, SupraFAB, Freie Universität Berlin, Altensteinstr. 23a, 14195 Berlin, Germany
| | - Pierre Parutto
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Scott Gratz
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Tanja Matkovic-Rachid
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Stefanie Ryglewski
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander M. Walter
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- Department of Neuroscience, University of Copenhagen, Copenhagen 2200, Denmark
| | - David Holcman
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Kate O‘Connor Giles
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Group Molecular Physiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Stephan J. Sigrist
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
18
|
Davis BA, Chen HY, Ye Z, Ostlund I, Tippani M, Das D, Sripathy SR, Wang Y, Martin JM, Shim G, Panchwagh NM, Moses RL, Farinelli F, Bohlen JF, Li M, Luikart BW, Jaffe AE, Maher BJ. TCF4 mutations disrupt synaptic function through dysregulation of RIMBP2 in patient-derived cortical neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524788. [PMID: 36712024 PMCID: PMC9882330 DOI: 10.1101/2023.01.19.524788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Genetic variation in the transcription factor 4 ( TCF4) gene is associated with risk for a variety of developmental and psychiatric conditions, which includes a syndromic form of ASD called Pitt Hopkins Syndrome (PTHS). TCF4 encodes an activity-dependent transcription factor that is highly expressed during cortical development and in animal models is shown to regulate various aspects of neuronal development and function. However, our understanding of how disease-causing mutations in TCF4 confer pathophysiology in a human context is lacking. Here we show that cortical neurons derived from patients with TCF4 mutations have deficits in spontaneous synaptic transmission, network excitability and homeostatic plasticity. Transcriptomic analysis indicates these phenotypes result from altered expression of genes involved in presynaptic neurotransmission and identifies the presynaptic binding protein, RIMBP2 as the most differentially expressed gene in PTHS neurons. Remarkably, TCF4-dependent deficits in spontaneous synaptic transmission and network excitability were rescued by increasing RIMBP2 expression in presynaptic neurons. Together, these results identify TCF4 as a critical transcriptional regulator of human synaptic development and plasticity and specifically identifies dysregulation of presynaptic function as an early pathophysiology in PTHS.
Collapse
Affiliation(s)
- Brittany A. Davis
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Huei-Ying Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Zengyou Ye
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Isaac Ostlund
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Debamitra Das
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Srinidhi Rao Sripathy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Yanhong Wang
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Jacqueline M. Martin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Gina Shim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Neel M. Panchwagh
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Rebecca L. Moses
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Federica Farinelli
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Joseph F. Bohlen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Meijie Li
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Bryan W. Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Andrew E. Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Brady J. Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
19
|
Mazille M, Buczak K, Scheiffele P, Mauger O. Stimulus-specific remodeling of the neuronal transcriptome through nuclear intron-retaining transcripts. EMBO J 2022; 41:e110192. [PMID: 36149731 DOI: 10.15252/embj.2021110192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 08/20/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
The nuclear envelope has long been considered primarily a physical barrier separating nuclear and cytosolic contents. More recently, nuclear compartmentalization has been shown to have additional regulatory functions in controlling gene expression. A sizeable proportion of protein-coding mRNAs is more prevalent in the nucleus than in the cytosol, suggesting regulated mRNA trafficking to the cytosol, but the mechanisms underlying controlled nuclear mRNA retention remain unclear. Here, we provide a comprehensive map of the subcellular localization of mRNAs in mature mouse cortical neurons, and reveal that transcripts retained in the nucleus comprise the majority of stable intron-retaining mRNAs. Systematically probing the fate of nuclear transcripts upon neuronal stimulation, we found opposite effects on sub-populations of transcripts: while some are targeted for degradation, others complete splicing to generate fully mature mRNAs that are exported to the cytosol and mediate rapid increases in protein levels. Finally, different forms of stimulation mobilize distinct groups of intron-retaining transcripts, with this selectivity arising from the activation of specific signaling pathways. Overall, our findings uncover a cue-specific control of intron retention as a major regulator of acute remodeling of the neuronal transcriptome.
Collapse
Affiliation(s)
- Maxime Mazille
- Biozentrum of the University of Basel, Basel, Switzerland
| | | | | | - Oriane Mauger
- Biozentrum of the University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
Sarkar D, Kang J, Wassie AT, Schroeder ME, Peng Z, Tarr TB, Tang AH, Niederst ED, Young JZ, Su H, Park D, Yin P, Tsai LH, Blanpied TA, Boyden ES. Revealing nanostructures in brain tissue via protein decrowding by iterative expansion microscopy. Nat Biomed Eng 2022; 6:1057-1073. [PMID: 36038771 PMCID: PMC9551354 DOI: 10.1038/s41551-022-00912-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
Many crowded biomolecular structures in cells and tissues are inaccessible to labelling antibodies. To understand how proteins within these structures are arranged with nanoscale precision therefore requires that these structures be decrowded before labelling. Here we show that an iterative variant of expansion microscopy (the permeation of cells and tissues by a swellable hydrogel followed by isotropic hydrogel expansion, to allow for enhanced imaging resolution with ordinary microscopes) enables the imaging of nanostructures in expanded yet otherwise intact tissues at a resolution of about 20 nm. The method, which we named 'expansion revealing' and validated with DNA-probe-based super-resolution microscopy, involves gel-anchoring reagents and the embedding, expansion and re-embedding of the sample in homogeneous swellable hydrogels. Expansion revealing enabled us to use confocal microscopy to image the alignment of pre-synaptic calcium channels with post-synaptic scaffolding proteins in intact brain circuits, and to uncover periodic amyloid nanoclusters containing ion-channel proteins in brain tissue from a mouse model of Alzheimer's disease. Expansion revealing will enable the further discovery of previously unseen nanostructures within cells and tissues.
Collapse
Affiliation(s)
- Deblina Sarkar
- Media Lab, MIT, Cambridge, MA, 02139, USA.,MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Jinyoung Kang
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Asmamaw T Wassie
- Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Margaret E. Schroeder
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA
| | - Zhuyu Peng
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Tyler B. Tarr
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, China
| | - Emily D. Niederst
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Jennie, Z. Young
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Hanquan Su
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Demian Park
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA. .,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Correspondence and requests for materials should be addressed to Li-Huei Tsai, Thomas A. Blanpied or Edward S. Boyden. , ,
| | - Edward S. Boyden
- MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, 02139, USA.,MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,Koch Institute, MIT, Cambridge, MA, 02139, USA.,Howard Hughes Medical Institute, Cambridge, MA, 02139, USA.,Media Arts and Sciences, MIT, Cambridge, MA, 02139, USA.,K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, 02139, USA.,Correspondence and requests for materials should be addressed to Li-Huei Tsai, Thomas A. Blanpied or Edward S. Boyden. , ,
| |
Collapse
|
21
|
Re-examination of the determinants of synaptic strength from the perspective of superresolution imaging. Curr Opin Neurobiol 2022; 74:102540. [DOI: 10.1016/j.conb.2022.102540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/04/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022]
|
22
|
Tan C, Wang SSH, de Nola G, Kaeser PS. Rebuilding essential active zone functions within a synapse. Neuron 2022; 110:1498-1515.e8. [PMID: 35176221 PMCID: PMC9081183 DOI: 10.1016/j.neuron.2022.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 01/15/2023]
Abstract
Presynaptic active zones are molecular machines that control neurotransmitter secretion. They form sites for vesicle docking and priming and couple vesicles to Ca2+ entry for release triggering. The complexity of active zone machinery has made it challenging to determine its mechanisms in release. Simultaneous knockout of the active zone proteins RIM and ELKS disrupts active zone assembly, abolishes vesicle docking, and impairs release. We here rebuild docking, priming, and Ca2+ secretion coupling in these mutants without reinstating active zone networks. Re-expression of RIM zinc fingers recruited Munc13 to undocked vesicles and rendered the vesicles release competent. Action potential triggering of release was reconstituted by docking these primed vesicles to Ca2+ channels through attaching RIM zinc fingers to CaVβ4-subunits. Our work identifies an 80-kDa β4-Zn protein that bypasses the need for megadalton-sized secretory machines, establishes that fusion competence and docking are mechanistically separable, and defines RIM zinc finger-Munc13 complexes as hubs for active zone function.
Collapse
Affiliation(s)
- Chao Tan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Shahoha M, Cohen R, Ben-Simon Y, Ashery U. cAMP-Dependent Synaptic Plasticity at the Hippocampal Mossy Fiber Terminal. Front Synaptic Neurosci 2022; 14:861215. [PMID: 35444523 PMCID: PMC9013808 DOI: 10.3389/fnsyn.2022.861215] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a crucial second messenger involved in both pre- and postsynaptic plasticity in many neuronal types across species. In the hippocampal mossy fiber (MF) synapse, cAMP mediates presynaptic long-term potentiation and depression. The main cAMP-dependent signaling pathway linked to MF synaptic plasticity acts via the activation of the protein kinase A (PKA) molecular cascade. Accordingly, various downstream putative synaptic PKA target proteins have been linked to cAMP-dependent MF synaptic plasticity, such as synapsin, rabphilin, synaptotagmin-12, RIM1a, tomosyn, and P/Q-type calcium channels. Regulating the expression of some of these proteins alters synaptic release probability and calcium channel clustering, resulting in short- and long-term changes to synaptic efficacy. However, despite decades of research, the exact molecular mechanisms by which cAMP and PKA exert their influences in MF terminals remain largely unknown. Here, we review current knowledge of different cAMP catalysts and potential downstream PKA-dependent molecular cascades, in addition to non-canonical cAMP-dependent but PKA-independent cascades, which might serve as alternative, compensatory or competing pathways to the canonical PKA cascade. Since several other central synapses share a similar form of presynaptic plasticity with the MF, a better description of the molecular mechanisms governing MF plasticity could be key to understanding the relationship between the transcriptional and computational levels across brain regions.
Collapse
Affiliation(s)
- Meishar Shahoha
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ronni Cohen
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Ben-Simon
- Department of Neurophysiology, Vienna Medical University, Vienna, Austria
- *Correspondence: Yoav Ben-Simon,
| | - Uri Ashery
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Uri Ashery,
| |
Collapse
|
24
|
Wang S, Ma C. Neuronal SNARE complex assembly guided by Munc18-1 and Munc13-1. FEBS Open Bio 2022; 12:1939-1957. [PMID: 35278279 PMCID: PMC9623535 DOI: 10.1002/2211-5463.13394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 01/25/2023] Open
Abstract
Neurotransmitter release by Ca2+ -triggered synaptic vesicle exocytosis is essential for information transmission in the nervous system. The soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs) syntaxin-1, SNAP-25, and synaptobrevin-2 form the SNARE complex to bring synaptic vesicles and the plasma membranes together and to catalyze membrane fusion. Munc18-1 and Munc13-1 regulate synaptic vesicle priming via orchestrating neuronal SNARE complex assembly. In this review, we summarize recent advances toward the functions and molecular mechanisms of Munc18-1 and Munc13-1 in guiding neuronal SNARE complex assembly, and discuss the functional similarities and differences between Munc18-1 and Munc13-1 in neurons and their homologs in other intracellular membrane trafficking systems.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
25
|
Holderith N, Aldahabi M, Nusser Z. Selective Enrichment of Munc13-2 in Presynaptic Active Zones of Hippocampal Pyramidal Cells That Innervate mGluR1α Expressing Interneurons. Front Synaptic Neurosci 2022; 13:773209. [PMID: 35221979 PMCID: PMC8866005 DOI: 10.3389/fnsyn.2021.773209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Selective distribution of proteins in presynaptic active zones (AZs) is a prerequisite for generating postsynaptic target cell type-specific differences in presynaptic vesicle release probability (Pv) and short-term plasticity, a characteristic feature of cortical pyramidal cells (PCs). In the hippocampus of rodents, somatostatin and mGluR1α expressing interneurons (mGluR1α+ INs) receive small, facilitating excitatory postsynaptic currents (EPSCs) from PCs and express Elfn1 that trans-synaptically recruits mGluR7 into the presynaptic AZ of PC axons. Here we show that Elfn1 also has a role in the selective recruitment of Munc13-2, a synaptic vesicle priming and docking protein, to PC AZs that innervate mGluR1α+ INs. In Elfn1 knock-out mice, unitary EPSCs (uEPSCs) in mGluR1α+ INs have threefold larger amplitudes with less pronounced short-term facilitation, which might be the consequence of the loss of either mGluR7 or Munc13-2 or both. Conditional genetic deletion of Munc13-2 from CA1 PCs results in the loss of Munc13-2, but not mGluR7 from the AZs, and has no effect on the amplitude of uEPSCs and leaves the characteristic short-term facilitation intact at PC to mGluR1α+ IN connection. Our results demonstrate that Munc13-1 alone is capable of imposing low Pv at PC to mGluR1α+ IN synapses and Munc13-2 has yet an unknown role in this synapse.
Collapse
Affiliation(s)
- Noemi Holderith
- Institute of Experimental Medicine, Eotvos Lorand Research Network, Budapest, Hungary
| | - Mohammad Aldahabi
- Institute of Experimental Medicine, Eotvos Lorand Research Network, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Zoltan Nusser
- Institute of Experimental Medicine, Eotvos Lorand Research Network, Budapest, Hungary
- *Correspondence: Zoltan Nusser,
| |
Collapse
|
26
|
Banerjee A, Imig C, Balakrishnan K, Kershberg L, Lipstein N, Uronen RL, Wang J, Cai X, Benseler F, Rhee JS, Cooper BH, Liu C, Wojcik SM, Brose N, Kaeser PS. Molecular and functional architecture of striatal dopamine release sites. Neuron 2022; 110:248-265.e9. [PMID: 34767769 PMCID: PMC8859508 DOI: 10.1016/j.neuron.2021.10.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 09/22/2021] [Accepted: 10/19/2021] [Indexed: 01/21/2023]
Abstract
Despite the importance of dopamine for striatal circuit function, mechanistic understanding of dopamine transmission remains incomplete. We recently showed that dopamine secretion relies on the presynaptic scaffolding protein RIM, indicating that it occurs at active zone-like sites similar to classical synaptic vesicle exocytosis. Here, we establish using a systematic gene knockout approach that Munc13 and Liprin-α, active zone proteins for vesicle priming and release site organization, are important for dopamine secretion. Furthermore, RIM zinc finger and C2B domains, which bind to Munc13 and Liprin-α, respectively, are needed to restore dopamine release after RIM ablation. In contrast, and different from typical synapses, the active zone scaffolds RIM-BP and ELKS, and RIM domains that bind to them, are expendable. Hence, dopamine release necessitates priming and release site scaffolding by RIM, Munc13, and Liprin-α, but other active zone proteins are dispensable. Our work establishes that efficient release site architecture mediates fast dopamine exocytosis.
Collapse
Affiliation(s)
- Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | | | - Lauren Kershberg
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Noa Lipstein
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Riikka-Liisa Uronen
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Jiexin Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Xintong Cai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Jeong Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Sonja M Wojcik
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Zhu X, Pan S, Li R, Chen Z, Xie X, Han D, Lv S, Huang Y. Novel Biomarker Genes for Prognosis of Survival and Treatment of Glioma. Front Oncol 2022; 11:667884. [PMID: 34976783 PMCID: PMC8714878 DOI: 10.3389/fonc.2021.667884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant primary central nervous system tumor. Although surgery, radiotherapy, and chemotherapy treatments are available, the 5-year survival rate of GBM is only 5.8%. Therefore, it is imperative to find novel biomarker for the prognosis and treatment of GBM. In this study, a total of 141 differentially expressed genes (DEGs) in GBM were identified by analyzing the GSE12657, GSE90886, and GSE90598 datasets. After reducing the data dimensionality, Kaplan-Meier survival analysis indicated that expression of PTPRN and RIM-BP2 were downregulated in GBM tissues when compared with that of normal tissues and that the expression of these genes was a good prognostic biomarker for GBM (p<0.05). Then, the GSE46531 dataset and the Genomics of Drug Sensitivity in Cancer (GDSC) database were used to examine the relationship between sensitivity radiotherapy (RT) and chemotherapy for GBM and expression of PTPRN and RIM-BP2. The expression of PTPRN was significantly high in RT-resistant patients (p<0.05) but it was not related to temozolomide (TMZ) resistance. The expression level of RIM-BP2 was not associated with RT or TMZ treatment. Among the chemotherapeutic drugs, cisplatin and erlotinib had a significantly good treatment effect for glioma with expression of PTPRN or RIM-BP2 and in lower-grade glioma (LGG) with IDH mutation. (p < 0.05). The tumor mutational burden (TMB) score in the low PTPRN expression group was significantly higher than that in the high PTPRN expression group (p=0.013), with a large degree of tumor immune cell infiltration. In conclusion, these findings contributed to the discovery process of potential biomarkers and therapeutic targets for glioma patients.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Sian Pan
- Department of Rehabilitation Medicine, Zhuzhou Central Hospital, Zhuzhou, China
| | - Rui Li
- Department of Operating Theatre, Zhuzhou Central Hospital, Zhuzhou, China
| | - Zebo Chen
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Xingyun Xie
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Deqing Han
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yongkai Huang
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| |
Collapse
|
28
|
Abstract
Fluorescence imaging techniques play a pivotal role in our understanding of the nervous system. The emergence of various super-resolution microscopy methods and specialized fluorescent probes enables direct insight into neuronal structure and protein arrangements in cellular subcompartments with so far unmatched resolution. Super-resolving visualization techniques in neurons unveil a novel understanding of cytoskeletal composition, distribution, motility, and signaling of membrane proteins, subsynaptic structure and function, and neuron-glia interaction. Well-defined molecular targets in autoimmune and neurodegenerative disease models provide excellent starting points for in-depth investigation of disease pathophysiology using novel and innovative imaging methodology. Application of super-resolution microscopy in human brain samples and for testing clinical biomarkers is still in its infancy but opens new opportunities for translational research in neurology and neuroscience. In this review, we describe how super-resolving microscopy has improved our understanding of neuronal and brain function and dysfunction in the last two decades.
Collapse
Affiliation(s)
- Christian Werner
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
29
|
Rapid Ca 2+ channel accumulation contributes to cAMP-mediated increase in transmission at hippocampal mossy fiber synapses. Proc Natl Acad Sci U S A 2021; 118:2016754118. [PMID: 33622791 DOI: 10.1073/pnas.2016754118] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The cyclic adenosine monophosphate (cAMP)-dependent potentiation of neurotransmitter release is important for higher brain functions such as learning and memory. To reveal the underlying mechanisms, we applied paired pre- and postsynaptic recordings from hippocampal mossy fiber-CA3 synapses. Ca2+ uncaging experiments did not reveal changes in the intracellular Ca2+ sensitivity for transmitter release by cAMP, but suggested an increase in the local Ca2+ concentration at the release site, which was much lower than that of other synapses before potentiation. Total internal reflection fluorescence (TIRF) microscopy indicated a clear increase in the local Ca2+ concentration at the release site within 5 to 10 min, suggesting that the increase in local Ca2+ is explained by the simple mechanism of rapid Ca2+ channel accumulation. Consistently, two-dimensional time-gated stimulated emission depletion microscopy (gSTED) microscopy showed an increase in the P/Q-type Ca2+ channel cluster size near the release sites. Taken together, this study suggests a potential mechanism for the cAMP-dependent increase in transmission at hippocampal mossy fiber-CA3 synapses, namely an accumulation of active zone Ca2+ channels.
Collapse
|
30
|
Orlando M, Dvorzhak A, Bruentgens F, Maglione M, Rost BR, Sigrist SJ, Breustedt J, Schmitz D. Recruitment of release sites underlies chemical presynaptic potentiation at hippocampal mossy fiber boutons. PLoS Biol 2021; 19:e3001149. [PMID: 34153028 PMCID: PMC8216508 DOI: 10.1371/journal.pbio.3001149] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/17/2021] [Indexed: 01/14/2023] Open
Abstract
Synaptic plasticity is a cellular model for learning and memory. However, the expression mechanisms underlying presynaptic forms of plasticity are not well understood. Here, we investigate functional and structural correlates of presynaptic potentiation at large hippocampal mossy fiber boutons induced by the adenylyl cyclase activator forskolin. We performed 2-photon imaging of the genetically encoded glutamate sensor iGluu that revealed an increase in the surface area used for glutamate release at potentiated terminals. Time-gated stimulated emission depletion microscopy revealed no change in the coupling distance between P/Q-type calcium channels and release sites mapped by Munc13-1 cluster position. Finally, by high-pressure freezing and transmission electron microscopy analysis, we found a fast remodeling of synaptic ultrastructure at potentiated boutons: Synaptic vesicles dispersed in the terminal and accumulated at the active zones, while active zone density and synaptic complexity increased. We suggest that these rapid and early structural rearrangements might enable long-term increase in synaptic strength.
Collapse
Affiliation(s)
- Marta Orlando
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Anton Dvorzhak
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Felicitas Bruentgens
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Marta Maglione
- NeuroCure Cluster of Excellence, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Benjamin R. Rost
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Stephan J. Sigrist
- NeuroCure Cluster of Excellence, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Jörg Breustedt
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Dietmar Schmitz
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
- German Center for Neurodegenerative Diseases, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
31
|
Emperador-Melero J, Wong MY, Wang SSH, de Nola G, Nyitrai H, Kirchhausen T, Kaeser PS. PKC-phosphorylation of Liprin-α3 triggers phase separation and controls presynaptic active zone structure. Nat Commun 2021; 12:3057. [PMID: 34031393 PMCID: PMC8144191 DOI: 10.1038/s41467-021-23116-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/16/2021] [Indexed: 01/24/2023] Open
Abstract
The active zone of a presynaptic nerve terminal defines sites for neurotransmitter release. Its protein machinery may be organized through liquid-liquid phase separation, a mechanism for the formation of membrane-less subcellular compartments. Here, we show that the active zone protein Liprin-α3 rapidly and reversibly undergoes phase separation in transfected HEK293T cells. Condensate formation is triggered by Liprin-α3 PKC-phosphorylation at serine-760, and RIM and Munc13 are co-recruited into membrane-attached condensates. Phospho-specific antibodies establish phosphorylation of Liprin-α3 serine-760 in transfected cells and mouse brain tissue. In primary hippocampal neurons of newly generated Liprin-α2/α3 double knockout mice, synaptic levels of RIM and Munc13 are reduced and the pool of releasable vesicles is decreased. Re-expression of Liprin-α3 restored these presynaptic defects, while mutating the Liprin-α3 phosphorylation site to abolish phase condensation prevented this rescue. Finally, PKC activation in these neurons acutely increased RIM, Munc13 and neurotransmitter release, which depended on the presence of phosphorylatable Liprin-α3. Our findings indicate that PKC-mediated phosphorylation of Liprin-α3 triggers its phase separation and modulates active zone structure and function.
Collapse
Affiliation(s)
| | - Man Yan Wong
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Hajnalka Nyitrai
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,VIB-KU Leuven Center for Brain and Disease Research, Campus Gasthuisberg, Leuven, Belgium
| | - Tom Kirchhausen
- Departments of Cell Biology and Pediatrics, Harvard Medical School and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
McDonald NA, Shen K. Finding functions of phase separation in the presynapse. Curr Opin Neurobiol 2021; 69:178-184. [PMID: 33979706 DOI: 10.1016/j.conb.2021.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/23/2022]
Abstract
Synapses are the basic units of neuronal communication. Understanding how synapses assemble and function is therefore essential to understanding nervous systems. Decades of study have identified many molecular components and functional mechanisms of synapses. Recently, an additional level of synaptic protein organization has been identified: phase separation. In the presynapse, components of the central active zone and a synaptic vesicle-clustering factor have been shown to form liquid-liquid phase-separated condensates or hydrogels. New in vivo functional studies have directly tested how phase separation impacts both synapse formation and function. Here, we review this emerging evidence for in vivo functional roles of phase separation at the presynapse and discuss future functional studies necessary to understand its complexity.
Collapse
Affiliation(s)
| | - Kang Shen
- Department of Biology, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
33
|
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
|
34
|
Mencacci NE, Brockmann MM, Dai J, Pajusalu S, Atasu B, Campos J, Pino G, Gonzalez-Latapi P, Patzke C, Schwake M, Tucci A, Pittman A, Simon-Sanchez J, Carvill GL, Balint B, Wiethoff S, Warner TT, Papandreou A, Soo A, Rein R, Kadastik-Eerme L, Puusepp S, Reinson K, Tomberg T, Hanagasi H, Gasser T, Bhatia KP, Kurian MA, Lohmann E, Õunap K, Rosenmund C, Südhof TC, Wood NW, Krainc D, Acuna C. Biallelic variants in TSPOAP1, encoding the active-zone protein RIMBP1, cause autosomal recessive dystonia. J Clin Invest 2021; 131:140625. [PMID: 33539324 DOI: 10.1172/jci140625] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/03/2021] [Indexed: 12/27/2022] Open
Abstract
Dystonia is a debilitating hyperkinetic movement disorder, which can be transmitted as a monogenic trait. Here, we describe homozygous frameshift, nonsense, and missense variants in TSPOAP1, which encodes the active-zone RIM-binding protein 1 (RIMBP1), as a genetic cause of autosomal recessive dystonia in 7 subjects from 3 unrelated families. Subjects carrying loss-of-function variants presented with juvenile-onset progressive generalized dystonia, associated with intellectual disability and cerebellar atrophy. Conversely, subjects carrying a pathogenic missense variant (p.Gly1808Ser) presented with isolated adult-onset focal dystonia. In mice, complete loss of RIMBP1, known to reduce neurotransmission, led to motor abnormalities reminiscent of dystonia, decreased Purkinje cell dendritic arborization, and reduced numbers of cerebellar synapses. In vitro analysis of the p.Gly1808Ser variant showed larger spike-evoked calcium transients and enhanced neurotransmission, suggesting that RIMBP1-linked dystonia can be caused by either reduced or enhanced rates of spike-evoked release in relevant neural networks. Our findings establish a direct link between dysfunction of the presynaptic active zone and dystonia and highlight the critical role played by well-balanced neurotransmission in motor control and disease pathogenesis.
Collapse
Affiliation(s)
- Niccolò E Mencacci
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Marisa M Brockmann
- Institute of Neurophysiology, Charité Universitätsmedizin, Berlin, Germany
| | - Jinye Dai
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Sander Pajusalu
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia.,Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Burcu Atasu
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany.,Center of Neurology, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Joaquin Campos
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, and
| | - Gabriela Pino
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, and
| | - Paulina Gonzalez-Latapi
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christopher Patzke
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Schwake
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Arianna Tucci
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Alan Pittman
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Javier Simon-Sanchez
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany.,Center of Neurology, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Gemma L Carvill
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bettina Balint
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sarah Wiethoff
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Center of Neurology, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Klinik für Neurologie mit Institut für Translationale Neurologie, Albert Schweitzer Campus 1, Gebäude A1, Münster, Germany
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Apostolos Papandreou
- Molecular Neurosciences, Developmental Neurosciences, UCL Institute of Child Health, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | - Audrey Soo
- Molecular Neurosciences, Developmental Neurosciences, UCL Institute of Child Health, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | | | | | - Sanna Puusepp
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia
| | - Karit Reinson
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia
| | - Tiiu Tomberg
- Radiology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Hasmet Hanagasi
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany.,Center of Neurology, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Manju A Kurian
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, United Kingdom.,Molecular Neurosciences, Developmental Neurosciences, UCL Institute of Child Health, London, United Kingdom
| | - Ebba Lohmann
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany.,Center of Neurology, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Katrin Õunap
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia
| | | | - Thomas C Südhof
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Nicholas W Wood
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Dimitri Krainc
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Claudio Acuna
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA.,Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, and
| |
Collapse
|
35
|
Pooryasin A, Maglione M, Schubert M, Matkovic-Rachid T, Hasheminasab SM, Pech U, Fiala A, Mielke T, Sigrist SJ. Unc13A and Unc13B contribute to the decoding of distinct sensory information in Drosophila. Nat Commun 2021; 12:1932. [PMID: 33771998 PMCID: PMC7997984 DOI: 10.1038/s41467-021-22180-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
The physical distance between presynaptic Ca2+ channels and the Ca2+ sensors triggering the release of neurotransmitter-containing vesicles regulates short-term plasticity (STP). While STP is highly diversified across synapse types, the computational and behavioral relevance of this diversity remains unclear. In the Drosophila brain, at nanoscale level, we can distinguish distinct coupling distances between Ca2+ channels and the (m)unc13 family priming factors, Unc13A and Unc13B. Importantly, coupling distance defines release components with distinct STP characteristics. Here, we show that while Unc13A and Unc13B both contribute to synaptic signalling, they play distinct roles in neural decoding of olfactory information at excitatory projection neuron (ePN) output synapses. Unc13A clusters closer to Ca2+ channels than Unc13B, specifically promoting fast phasic signal transfer. Reduction of Unc13A in ePNs attenuates responses to both aversive and appetitive stimuli, while reduction of Unc13B provokes a general shift towards appetitive values. Collectively, we provide direct genetic evidence that release components of distinct nanoscopic coupling distances differentially control STP to play distinct roles in neural decoding of sensory information. The physical distance between synaptic Ca2+ channels and sensors modulates short-term plasticity (STP). Here, the authors show that synaptic release factors Unc13A and Unc13B distinctly couple with Ca2+ channels and contribute to the neural decoding of distinct sensory information in Drosophila.
Collapse
Affiliation(s)
- Atefeh Pooryasin
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Marta Maglione
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Marco Schubert
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | | | - Sayed-Mohammad Hasheminasab
- Department of Dermatology, Venereology and Allergology, Charité Universitätsmedizin, Berlin, Germany.,CCU Translational Radiation Oncology, DKTK, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Pech
- Department of Molecular Neurobiology of Behavior, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, University of Göttingen, Göttingen, Germany.,Laboratory of Neuronal Communication, VIB Center for the Biology of Disease, K.U.Leuven, Leuven, Belgium
| | - André Fiala
- Department of Molecular Neurobiology of Behavior, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| | - Thorsten Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Group, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
36
|
Petzoldt AG, Götz TWB, Driller JH, Lützkendorf J, Reddy-Alla S, Matkovic-Rachid T, Liu S, Knoche E, Mertel S, Ugorets V, Lehmann M, Ramesh N, Beuschel CB, Kuropka B, Freund C, Stelzl U, Loll B, Liu F, Wahl MC, Sigrist SJ. RIM-binding protein couples synaptic vesicle recruitment to release sites. J Cell Biol 2021; 219:151735. [PMID: 32369542 PMCID: PMC7337501 DOI: 10.1083/jcb.201902059] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 02/03/2020] [Accepted: 04/07/2020] [Indexed: 11/24/2022] Open
Abstract
At presynaptic active zones, arrays of large conserved scaffold proteins mediate fast and temporally precise release of synaptic vesicles (SVs). SV release sites could be identified by clusters of Munc13, which allow SVs to dock in defined nanoscale relation to Ca2+ channels. We here show in Drosophila that RIM-binding protein (RIM-BP) connects release sites physically and functionally to the ELKS family Bruchpilot (BRP)-based scaffold engaged in SV recruitment. The RIM-BP N-terminal domain, while dispensable for SV release site organization, was crucial for proper nanoscale patterning of the BRP scaffold and needed for SV recruitment of SVs under strong stimulation. Structural analysis further showed that the RIM-BP fibronectin domains form a “hinge” in the protein center, while the C-terminal SH3 domain tandem binds RIM, Munc13, and Ca2+ channels release machinery collectively. RIM-BPs’ conserved domain architecture seemingly provides a relay to guide SVs from membrane far scaffolds into membrane close release sites.
Collapse
Affiliation(s)
- Astrid G Petzoldt
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - Torsten W B Götz
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - Jan Heiner Driller
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry Berlin, Berlin, Germany
| | - Janine Lützkendorf
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - Suneel Reddy-Alla
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | | | - Sunbin Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry Berlin, Berlin, Germany
| | - Elena Knoche
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - Sara Mertel
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - Vladimir Ugorets
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V., Campus Berlin-Buch, Berlin, Germany
| | - Niraja Ramesh
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | | | - Benno Kuropka
- Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Christian Freund
- Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Ulrich Stelzl
- Institut für Pharmazeutische Wissenschaften, Graz, Austria
| | - Bernhard Loll
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry Berlin, Berlin, Germany
| | - Fan Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V., Campus Berlin-Buch, Berlin, Germany
| | - Markus C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry Berlin, Berlin, Germany.,Helmholtz-Zentrum Berlin für Materialien und Energie, Macromolecular Crystallography, Berlin, Germany
| | - Stephan J Sigrist
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany.,NeuroCure, Charité, Berlin, Germany
| |
Collapse
|
37
|
Gao T, Zhang Z, Yang Y, Zhang H, Li N, Liu B. Impact of RIM-BPs in neuronal vesicles release. Brain Res Bull 2021; 170:129-136. [PMID: 33581313 DOI: 10.1016/j.brainresbull.2021.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Accurate signal transmission between neurons is accomplished by vesicle release with high spatiotemporal resolution in the central nervous system. The vesicle release occurs mainly in the active zone (AZ), a unique area on the presynaptic membrane. Many structural proteins expressed in the AZ connect with other proteins nearby. They can also regulate the precise release of vesicles through protein-protein interactions. RIM-binding proteins (RIM-BPs) are one of the essential proteins in the AZ. This review summarizes the structures and functions of three subtypes of RIM-BPs, including the interaction between RIM-BPs and other proteins such as Bassoon and voltage-gated calcium channel, their significance in stabilizing the AZ structure in the presynaptic region and collecting ion channels, and ultimately regulating the fusion and release of neuronal vesicles.
Collapse
Affiliation(s)
- Tianyu Gao
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Panjin Campus of Dalian University of Technology, Panjin, 124221, China
| | - Yunong Yang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China
| | - Na Li
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China.
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
38
|
Imbalanced post- and extrasynaptic SHANK2A functions during development affect social behavior in SHANK2-mediated neuropsychiatric disorders. Mol Psychiatry 2021; 26:6482-6504. [PMID: 34021263 PMCID: PMC8760046 DOI: 10.1038/s41380-021-01140-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Mutations in SHANK genes play an undisputed role in neuropsychiatric disorders. Until now, research has focused on the postsynaptic function of SHANKs, and prominent postsynaptic alterations in glutamatergic signal transmission have been reported in Shank KO mouse models. Recent studies have also suggested a possible presynaptic function of SHANK proteins, but these remain poorly defined. In this study, we examined how SHANK2 can mediate electrophysiological, molecular, and behavioral effects by conditionally overexpressing either wild-type SHANK2A or the extrasynaptic SHANK2A(R462X) variant. SHANK2A overexpression affected pre- and postsynaptic targets and revealed a reversible, development-dependent autism spectrum disorder-like behavior. SHANK2A also mediated redistribution of Ca2+-permeable AMPA receptors between apical and basal hippocampal CA1 dendrites, leading to impaired synaptic plasticity in the basal dendrites. Moreover, SHANK2A overexpression reduced social interaction and increased the excitatory noise in the olfactory cortex during odor processing. In contrast, overexpression of the extrasynaptic SHANK2A(R462X) variant did not impair hippocampal synaptic plasticity, but still altered the expression of presynaptic/axonal signaling proteins. We also observed an attention-deficit/hyperactivity-like behavior and improved social interaction along with enhanced signal-to-noise ratio in cortical odor processing. Our results suggest that the disruption of pre- and postsynaptic SHANK2 functions caused by SHANK2 mutations has a strong impact on social behavior. These findings indicate that pre- and postsynaptic SHANK2 actions cooperate for normal neuronal function, and that an imbalance between these functions may lead to different neuropsychiatric disorders.
Collapse
|
39
|
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
|
40
|
Coppola U, Olivo P, D’Aniello E, Johnson CJ, Stolfi A, Ristoratore F. Rimbp, a New Marker for the Nervous System of the Tunicate Ciona robusta. Genes (Basel) 2020; 11:genes11091006. [PMID: 32867148 PMCID: PMC7565545 DOI: 10.3390/genes11091006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/16/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Establishment of presynaptic mechanisms by proteins that regulate neurotransmitter release in the presynaptic active zone is considered a fundamental step in animal evolution. Rab3 interacting molecule-binding proteins (Rimbps) are crucial components of the presynaptic active zone and key players in calcium homeostasis. Although Rimbp involvement in these dynamics has been described in distantly related models such as fly and human, the role of this family in most invertebrates remains obscure. To fill this gap, we defined the evolutionary history of Rimbp family in animals, from sponges to mammals. We report, for the first time, the expression of the two isoforms of the unique Rimbp family member in Ciona robusta in distinct domains of the larval nervous system. We identify intronic enhancers that are able to drive expression in different nervous system territories partially corresponding to Rimbp endogenous expression. The analysis of gene expression patterns and the identification of regulatory elements of Rimbp will positively impact our understanding of this family of genes in the context of Ciona embryogenesis.
Collapse
Affiliation(s)
- Ugo Coppola
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (U.C.); (P.O.); (E.D.)
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Paola Olivo
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (U.C.); (P.O.); (E.D.)
| | - Enrico D’Aniello
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (U.C.); (P.O.); (E.D.)
| | | | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- Correspondence: (A.S.); (F.R.)
| | - Filomena Ristoratore
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (U.C.); (P.O.); (E.D.)
- Correspondence: (A.S.); (F.R.)
| |
Collapse
|
41
|
Vardar G, Gerth F, Schmitt XJ, Rautenstrauch P, Trimbuch T, Schubert J, Lerche H, Rosenmund C, Freund C. Epilepsy-causing STX1B mutations translate altered protein functions into distinct phenotypes in mouse neurons. Brain 2020; 143:2119-2138. [DOI: 10.1093/brain/awaa151] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/05/2020] [Accepted: 03/25/2020] [Indexed: 01/21/2023] Open
Abstract
AbstractSyntaxin 1B (STX1B) is a core component of the N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex that is critical for the exocytosis of synaptic vesicles in the presynapse. SNARE-mediated vesicle fusion is assisted by Munc18-1, which recruits STX1B in the auto-inhibited conformation, while Munc13 catalyses the fast and efficient pairing of helices during SNARE complex formation. Mutations within the STX1B gene are associated with epilepsy. Here we analysed three STX1B mutations by biochemical and electrophysiological means. These three paradigmatic mutations cause epilepsy syndromes of different severity, from benign fever-associated seizures in childhood to severe epileptic encephalopathies. An insertion/deletion (K45/RMCIE, L46M) mutation (STX1BInDel), causing mild epilepsy and located in the early helical Habc domain, leads to an unfolded protein unable to sustain neurotransmission. STX1BG226R, causing epileptic encephalopathies, strongly compromises the interaction with Munc18-1 and reduces expression of both proteins, the size of the readily releasable pool of vesicles, and Ca2+-triggered neurotransmitter release when expressed in STX1-null neurons. The mutation STX1BV216E, also causing epileptic encephalopathies, only slightly diminishes Munc18-1 and Munc13 interactions, but leads to enhanced fusogenicity and increased vesicular release probability, also in STX1-null neurons. Even though the synaptic output remained unchanged in excitatory hippocampal STX1B+/− neurons exogenously expressing STX1B mutants, the manifestation of clear and distinct molecular disease mechanisms by these mutants suggest that certain forms of epilepsies can be conceptualized by assigning mutations to structurally sensitive regions of the STX1B−Munc18-1 interface, translating into distinct neurophysiological phenotypes.
Collapse
Affiliation(s)
- Gülçin Vardar
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Germany
| | - Fabian Gerth
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Xiao Jakob Schmitt
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Pia Rautenstrauch
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Germany
| | - Julian Schubert
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | | | - Christian Freund
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| |
Collapse
|
42
|
Assembly of the presynaptic active zone. Curr Opin Neurobiol 2020; 63:95-103. [PMID: 32403081 DOI: 10.1016/j.conb.2020.03.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/29/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022]
Abstract
In a presynaptic nerve terminal, the active zone is composed of sophisticated protein machinery that enables secretion on a submillisecond time scale and precisely targets it toward postsynaptic receptors. The past two decades have provided deep insight into the roles of active zone proteins in exocytosis, but we are only beginning to understand how a neuron assembles active zone protein complexes into effective molecular machines. In this review, we outline the fundamental processes that are necessary for active zone assembly and discuss recent advances in understanding assembly mechanisms that arise from genetic, morphological and biochemical studies. We further outline the challenges ahead for understanding this important problem.
Collapse
|
43
|
Nanoscale Location Matters: Emerging Principles of Ca 2+ Channel Organization at the Presynaptic Active Zone. Neuron 2020; 104:627-629. [PMID: 31751541 DOI: 10.1016/j.neuron.2019.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
How does diversity in the organization of secretory machines determine properties of neurotransmitter release? In this issue of Neuron, Rebola et al. (2019) found that distinct nanoscale assemblies of Ca2+ channels and Munc13, not overall channel abundance, mediate differing release characteristics of two cerebellar synapses.
Collapse
|
44
|
Heine M, Holcman D. Asymmetry Between Pre- and Postsynaptic Transient Nanodomains Shapes Neuronal Communication. Trends Neurosci 2020; 43:182-196. [DOI: 10.1016/j.tins.2020.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022]
|