1
|
Kim H, Melliti N, Breithausen E, Michel K, Colomer SF, Poguzhelskaya E, Nemcova P, Ewell L, Blaess S, Becker A, Pitsch J, Dietrich D, Schoch S. Paroxysmal dystonia results from the loss of RIM4 in Purkinje cells. Brain 2024; 147:3171-3188. [PMID: 38478593 DOI: 10.1093/brain/awae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 09/04/2024] Open
Abstract
Full-length RIM1 and 2 are key components of the presynaptic active zone that ubiquitously control excitatory and inhibitory neurotransmitter release. Here, we report that the function of the small RIM isoform RIM4, consisting of a single C2 domain, is strikingly different from that of the long isoforms. RIM4 is dispensable for neurotransmitter release but plays a postsynaptic, cell type-specific role in cerebellar Purkinje cells that is essential for normal motor function. In the absence of RIM4, Purkinje cell intrinsic firing is reduced and caffeine-sensitive, and dendritic integration of climbing fibre input is disturbed. Mice lacking RIM4, but not mice lacking RIM1/2, selectively in Purkinje cells exhibit a severe, hours-long paroxysmal dystonia. These episodes can also be induced by caffeine, ethanol or stress and closely resemble the deficits seen with mutations of the PNKD (paroxysmal non-kinesigenic dystonia) gene. Our data reveal essential postsynaptic functions of RIM proteins and show non-overlapping specialized functions of a small isoform despite high homology to a single domain in the full-length proteins.
Collapse
Affiliation(s)
- Hyuntae Kim
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Nesrine Melliti
- Synaptic Neuroscience Team, Institute of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Eva Breithausen
- Synaptic Neuroscience Team, Institute of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Katrin Michel
- Synaptic Neuroscience Team, Institute of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Sara Ferrando Colomer
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Ekaterina Poguzhelskaya
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Paulina Nemcova
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Laura Ewell
- School of Medicine, UC Irvine, 92697 Irvine, USA
| | - Sandra Blaess
- Institute of Reconstructive Neurobiology, University Hospital Bonn, 53127 Bonn, Germany
| | - Albert Becker
- Synaptic Neuroscience Team, Institute of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Julika Pitsch
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Dirk Dietrich
- Synaptic Neuroscience Team, Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Susanne Schoch
- Synaptic Neuroscience Team, Institute of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
2
|
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 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
|
3
|
Laighneach A, Kelly JP, Desbonnet L, Holleran L, Kerr DM, McKernan D, Donohoe G, Morris DW. Social isolation-induced transcriptomic changes in mouse hippocampus impact the synapse and show convergence with human genetic risk for neurodevelopmental phenotypes. PLoS One 2023; 18:e0295855. [PMID: 38127959 PMCID: PMC10735045 DOI: 10.1371/journal.pone.0295855] [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: 09/11/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Early life stress (ELS) can impact brain development and is a risk factor for neurodevelopmental disorders such as schizophrenia. Post-weaning social isolation (SI) is used to model ELS in animals, using isolation stress to disrupt a normal developmental trajectory. We aimed to investigate how SI affects the expression of genes in mouse hippocampus and to investigate how these changes related to the genetic basis of neurodevelopmental phenotypes. BL/6J mice were exposed to post-weaning SI (PD21-25) or treated as group-housed controls (n = 7-8 per group). RNA sequencing was performed on tissue samples from the hippocampus of adult male and female mice. Four hundred and 1,215 differentially-expressed genes (DEGs) at a false discovery rate of < 0.05 were detected between SI and control samples for males and females respectively. DEGS for both males and females were significantly overrepresented in gene ontologies related to synaptic structure and function, especially the post-synapse. DEGs were enriched for common variant (SNP) heritability in humans that contributes to risk of neuropsychiatric disorders (schizophrenia, bipolar disorder) and to cognitive function. DEGs were also enriched for genes harbouring rare de novo variants that contribute to autism spectrum disorder and other developmental disorders. Finally, cell type analysis revealed populations of hippocampal astrocytes that were enriched for DEGs, indicating effects in these cell types as well as neurons. Overall, these data suggest a convergence between genes dysregulated by the SI stressor in the mouse and genes associated with neurodevelopmental disorders and cognitive phenotypes in humans.
Collapse
Affiliation(s)
- Aodán Laighneach
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| | - John P. Kelly
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Lieve Desbonnet
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Laurena Holleran
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| | - Daniel M. Kerr
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Declan McKernan
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| | - Derek W. Morris
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| |
Collapse
|
4
|
Mrestani A, Dannhäuser S, Pauli M, Kollmannsberger P, Hübsch M, Morris L, Langenhan T, Heckmann M, Paul MM. Nanoscaled RIM clustering at presynaptic active zones revealed by endogenous tagging. Life Sci Alliance 2023; 6:e202302021. [PMID: 37696575 PMCID: PMC10494931 DOI: 10.26508/lsa.202302021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
Chemical synaptic transmission involves neurotransmitter release from presynaptic active zones (AZs). The AZ protein Rab-3-interacting molecule (RIM) is important for normal Ca2+-triggered release. However, its precise localization within AZs of the glutamatergic neuromuscular junctions of Drosophila melanogaster remains elusive. We used CRISPR/Cas9-assisted genome engineering of the rim locus to incorporate small epitope tags for targeted super-resolution imaging. A V5-tag, derived from simian virus 5, and an HA-tag, derived from human influenza virus, were N-terminally fused to the RIM Zinc finger. Whereas both variants are expressed in co-localization with the core AZ scaffold Bruchpilot, electrophysiological characterization reveals that AP-evoked synaptic release is disturbed in rimV5-Znf but not in rimHA-Znf In addition, rimHA-Znf synapses show intact presynaptic homeostatic potentiation. Combining super-resolution localization microscopy and hierarchical clustering, we detect ∼10 RIMHA-Znf subclusters with ∼13 nm diameter per AZ that are compacted and increased in numbers in presynaptic homeostatic potentiation.
Collapse
Affiliation(s)
- Achmed Mrestani
- https://ror.org/00fbnyb24 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Neurology, Leipzig University Medical Center, Leipzig, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Sven Dannhäuser
- https://ror.org/00fbnyb24 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Martin Pauli
- https://ror.org/00fbnyb24 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Martha Hübsch
- https://ror.org/00fbnyb24 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lydia Morris
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Tobias Langenhan
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Manfred Heckmann
- https://ror.org/00fbnyb24 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Mila M Paul
- https://ror.org/00fbnyb24 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- https://ror.org/03pvr2g57 Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
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: 2.0] [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
|
6
|
Mizrahi L, Choudhary A, Ofer P, Goldberg G, Milanesi E, Kelsoe JR, Gurwitz D, Alda M, Gage FH, Stern S. Immunoglobulin genes expressed in lymphoblastoid cell lines discern and predict lithium response in bipolar disorder patients. Mol Psychiatry 2023; 28:4280-4293. [PMID: 37488168 PMCID: PMC10827667 DOI: 10.1038/s41380-023-02183-z] [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: 09/13/2022] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023]
Abstract
Bipolar disorder (BD) is a neuropsychiatric mood disorder manifested by recurrent episodes of mania and depression. More than half of BD patients are non-responsive to lithium, the first-line treatment drug, complicating BD clinical management. Given its unknown etiology, it is pertinent to understand the genetic signatures that lead to variability in lithium response. We discovered a set of differentially expressed genes (DEGs) from the lymphoblastoid cell lines (LCLs) of 10 controls and 19 BD patients belonging mainly to the immunoglobulin gene family that can be used as potential biomarkers to diagnose and treat BD. Importantly, we trained machine learning algorithms on our datasets that predicted the lithium response of BD subtypes with minimal errors, even when used on a different cohort of 24 BD patients acquired by a different laboratory. This proves the scalability of our methodology for predicting lithium response in BD and for a prompt and suitable decision on therapeutic interventions.
Collapse
Affiliation(s)
- Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3498838, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3498838, Israel
| | - Polina Ofer
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3498838, Israel
| | - Gabriela Goldberg
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Elena Milanesi
- Victor Babes National Institute of Pathology, Bucharest, 050096, Romania
| | - John R Kelsoe
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, B3H 2E2, Canada
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
7
|
Navarro-Martínez A, Vicente-García C, Carvajal JJ. NMJ-related diseases beyond the congenital myasthenic syndromes. Front Cell Dev Biol 2023; 11:1216726. [PMID: 37601107 PMCID: PMC10436495 DOI: 10.3389/fcell.2023.1216726] [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: 05/04/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Neuromuscular junctions (NMJs) are a special type of chemical synapse that transmits electrical stimuli from motor neurons (MNs) to their innervating skeletal muscle to induce a motor response. They are an ideal model for the study of synapses, given their manageable size and easy accessibility. Alterations in their morphology or function lead to neuromuscular disorders, such as the congenital myasthenic syndromes, which are caused by mutations in proteins located in the NMJ. In this review, we highlight novel potential candidate genes that may cause or modify NMJs-related pathologies in humans by exploring the phenotypes of hundreds of mouse models available in the literature. We also underscore the fact that NMJs may differ between species, muscles or even sexes. Hence the importance of choosing a good model organism for the study of NMJ-related diseases: only taking into account the specific features of the mammalian NMJ, experimental results would be efficiently translated to the clinic.
Collapse
Affiliation(s)
| | - Cristina Vicente-García
- Centro Andaluz de Biología del Desarrollo, CSIC-UPO-JA, Universidad Pablo de Olavide, Sevilla, Spain
| | | |
Collapse
|
8
|
Oh KH, Xiong A, Choe JY, Richmond JE, Kim H. Active Zone Trafficking of CaV2/UNC-2 Channels Is Independent of β/CCB-1 and α2δ/UNC-36 Subunits. J Neurosci 2023; 43:5142-5157. [PMID: 37160370 PMCID: PMC10343168 DOI: 10.1523/jneurosci.2264-22.2023] [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: 12/09/2022] [Revised: 03/28/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023] Open
Abstract
The CaV2 voltage-gated calcium channel is the major conduit of calcium ions necessary for neurotransmitter release at presynaptic active zones (AZs). The CaV2 channel is a multimeric complex that consists of a pore-forming α1 subunit and two auxiliary β and α2δ subunits. Although auxiliary subunits are critical for channel function, whether they are required for α1 trafficking is unresolved. Using endogenously fluorescent protein-tagged CaV2 channel subunits in Caenorhabditis elegans, we show that UNC-2/α1 localizes to AZs even in the absence of CCB-1/β or UNC-36/α2δ, albeit at low levels. When UNC-2 is manipulated to be trapped in the endoplasmic reticulum (ER), CCB-1 and UNC-36 fail to colocalize with UNC-2 in the ER, indicating that they do not coassemble with UNC-2 in the ER. Moreover, blocking ER-associated degradation does not further increase presynaptic UNC-2 channels in ccb-1 or unc-36 mutants, indicating that UNC-2 levels are not regulated in the ER. An unc-2 mutant lacking C-terminal AZ protein interaction sites with intact auxiliary subunit binding sites displays persistent presynaptic UNC-2 localization and a prominent increase of UNC-2 channels in nonsynaptic axonal regions, underscoring a protective role of auxiliary subunits against UNC-2 degradation. In the absence of UNC-2, presynaptic CCB-1 and UNC-36 are profoundly diminished to barely detectable levels, indicating that UNC-2 is required for the presynaptic localization of CCB-1 and UNC-36. Together, our findings demonstrate that although the pore-forming subunit does not require auxiliary subunits for its trafficking and transport to AZs, it recruits auxiliary subunits to stabilize and expand calcium channel signalosomes.SIGNIFICANCE STATEMENT Synaptic transmission in the neuron hinges on the coupling of synaptic vesicle exocytosis with calcium influx. This calcium influx is mediated by CaV2 voltage-gated calcium channels. These channels consist of one pore-forming α1 subunit and two auxiliary β and α2δ subunits. The auxiliary subunits enhance channel function and regulate the overall level of channels at presynaptic terminals. However, it is not settled how these auxiliary subunits regulate the overall channel level. Our study in C. elegans finds that although the auxiliary subunits do not coassemble with α1 and aid trafficking, they are recruited to α1 and stabilize the channel complex at presynaptic terminals. Our study suggests that drugs that target the auxiliary subunits can directly destabilize and have an impact on CaV2 channels.
Collapse
Affiliation(s)
- Kelly H Oh
- Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Ame Xiong
- Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Jun-Yong Choe
- Department of Chemistry, East Carolina University, Greenville, North Carolina 27858
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois, Chicago, Illinois 60607
| | - Hongkyun Kim
- Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| |
Collapse
|
9
|
Wu S, Fan J, Tang F, Chen L, Zhang X, Xiao D, Li X. The role of RIM in neurotransmitter release: promotion of synaptic vesicle docking, priming, and fusion. Front Neurosci 2023; 17:1123561. [PMID: 37179554 PMCID: PMC10169678 DOI: 10.3389/fnins.2023.1123561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
There are many special sites at the end of a synapse called active zones (AZs). Synaptic vesicles (SVs) fuse with presynaptic membranes at these sites, and this fusion is an important step in neurotransmitter release. The cytomatrix in the active zone (CAZ) is made up of proteins such as the regulating synaptic membrane exocytosis protein (RIM), RIM-binding proteins (RIM-BPs), ELKS/CAST, Bassoon/Piccolo, Liprin-α, and Munc13-1. RIM is a scaffold protein that interacts with CAZ proteins and presynaptic functional components to affect the docking, priming, and fusion of SVs. RIM is believed to play an important role in regulating the release of neurotransmitters (NTs). In addition, abnormal expression of RIM has been detected in many diseases, such as retinal diseases, Asperger's syndrome (AS), and degenerative scoliosis. Therefore, we believe that studying the molecular structure of RIM and its role in neurotransmitter release will help to clarify the molecular mechanism of neurotransmitter release and identify targets for the diagnosis and treatment of the aforementioned diseases.
Collapse
Affiliation(s)
- Shanshan Wu
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Jiali Fan
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Fajuan Tang
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Lin Chen
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaoyan Zhang
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Dongqiong Xiao
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xihong Li
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
10
|
Seitter H, Obkircher J, Grabher P, Hartl J, Zanetti L, Lux UT, Fotakis G, Fernández-Quintero ML, Kaserer T, Koschak A. A novel calcium channel Cavβ 2 splice variant with unique properties predominates in the retina. J Biol Chem 2023; 299:102972. [PMID: 36738788 PMCID: PMC10074810 DOI: 10.1016/j.jbc.2023.102972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Cavβ subunits are essential for surface expression of voltage-gated calcium channel complexes and crucially modulate biophysical properties like voltage-dependent inactivation. Here, we describe the discovery and characterization of a novel Cavβ2 variant with distinct features that predominates in the retina. We determined spliced exons in retinal transcripts of the Cacnb2 gene, coding for Cavβ2, by RNA-Seq data analysis and quantitative PCR. We cloned a novel Cavβ2 splice variant from mouse retina, which we are calling β2i, and investigated biophysical properties of calcium currents with this variant in a heterologous expression system as well as its intrinsic membrane interaction when expressed alone. Our data showed that β2i predominated in the retina with expression in photoreceptors and bipolar cells. Furthermore, we observed that the β2i N-terminus exhibited an extraordinary concentration of hydrophobic residues, a distinct feature not seen in canonical variants. The biophysical properties resembled known membrane-associated variants, and β2i exhibited both a strong membrane association and a propensity for clustering, which depended on hydrophobic residues in its N-terminus. We considered available Cavβ structure data to elucidate potential mechanisms underlying the observed characteristics but resolved N-terminus structures were lacking and thus, precluded clear conclusions. With this description of a novel N-terminus variant of Cavβ2, we expand the scope of functional variation through N-terminal splicing with a distinct form of membrane attachment. Further investigation of the molecular mechanisms underlying the features of β2i could provide new angles on the way Cavβ subunits modulate Ca2+ channels at the plasma membrane.
Collapse
Affiliation(s)
- Hartwig Seitter
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria.
| | - Jana Obkircher
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Patricia Grabher
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Julia Hartl
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Lucia Zanetti
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Uwe Thorsten Lux
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georgios Fotakis
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Teresa Kaserer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
11
|
Cunningham KL, Littleton JT. Mechanisms controlling the trafficking, localization, and abundance of presynaptic Ca 2+ channels. Front Mol Neurosci 2023; 15:1116729. [PMID: 36710932 PMCID: PMC9880069 DOI: 10.3389/fnmol.2022.1116729] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 01/14/2023] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) mediate Ca2+ influx to trigger neurotransmitter release at specialized presynaptic sites termed active zones (AZs). The abundance of VGCCs at AZs regulates neurotransmitter release probability (Pr ), a key presynaptic determinant of synaptic strength. Given this functional significance, defining the processes that cooperate to establish AZ VGCC abundance is critical for understanding how these mechanisms set synaptic strength and how they might be regulated to control presynaptic plasticity. VGCC abundance at AZs involves multiple steps, including channel biosynthesis (transcription, translation, and trafficking through the endomembrane system), forward axonal trafficking and delivery to synaptic terminals, incorporation and retention at presynaptic sites, and protein recycling. Here we discuss mechanisms that control VGCC abundance at synapses, highlighting findings from invertebrate and vertebrate models.
Collapse
Affiliation(s)
- Karen L. Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | |
Collapse
|
12
|
Zong P, Yue L. Regulation of Presynaptic Calcium Channels. ADVANCES IN NEUROBIOLOGY 2023; 33:171-202. [PMID: 37615867 DOI: 10.1007/978-3-031-34229-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Voltage-gated calcium channels (VGCCs), especially Cav2.1 and Cav2.2, are the major mediators of Ca2+ influx at the presynaptic membrane in response to neuron excitation, thereby exerting a predominant control on synaptic transmission. To guarantee the timely and precise release of neurotransmitters at synapses, the activity of presynaptic VGCCs is tightly regulated by a variety of factors, including auxiliary subunits, membrane potential, G protein-coupled receptors (GPCRs), calmodulin (CaM), Ca2+-binding proteins (CaBP), protein kinases, various interacting proteins, alternative splicing events, and genetic variations.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
13
|
Midorikawa M. Developmental and activity-dependent modulation of coupling distance between release site and Ca2+ channel. Front Cell Neurosci 2022; 16:1037721. [DOI: 10.3389/fncel.2022.1037721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Synapses are junctions between a presynaptic neuron and a postsynaptic cell specialized for fast and precise information transfer. The presynaptic terminal secretes neurotransmitters via exocytosis of synaptic vesicles. Exocytosis is a tightly regulated reaction that occurs within a millisecond of the arrival of an action potential. One crucial parameter in determining the characteristics of the transmitter release kinetics is the coupling distance between the release site and the Ca2+ channel. Still, the technical limitations have hindered detailed analysis from addressing how the coupling distance is regulated depending on the development or activity of the synapse. However, recent technical advances in electrophysiology and imaging are unveiling their different configurations in different conditions. Here, I will summarize developmental- and activity-dependent changes in the coupling distances revealed by recent studies.
Collapse
|
14
|
Lichter K, Paul MM, Pauli M, Schoch S, Kollmannsberger P, Stigloher C, Heckmann M, Sirén AL. Ultrastructural analysis of wild-type and RIM1α knockout active zones in a large cortical synapse. Cell Rep 2022; 40:111382. [PMID: 36130490 DOI: 10.1016/j.celrep.2022.111382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/14/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022] Open
Abstract
Rab3A-interacting molecule (RIM) is crucial for fast Ca2+-triggered synaptic vesicle (SV) release in presynaptic active zones (AZs). We investigated hippocampal giant mossy fiber bouton (MFB) AZ architecture in 3D using electron tomography of rapid cryo-immobilized acute brain slices in RIM1α-/- and wild-type mice. In RIM1α-/-, AZs are larger with increased synaptic cleft widths and a 3-fold reduced number of tightly docked SVs (0-2 nm). The distance of tightly docked SVs to the AZ center is increased from 110 to 195 nm, and the width of their electron-dense material between outer SV membrane and AZ membrane is reduced. Furthermore, the SV pool in RIM1α-/- is more heterogeneous. Thus, RIM1α, besides its role in tight SV docking, is crucial for synaptic architecture and vesicle pool organization in MFBs.
Collapse
Affiliation(s)
- Katharina Lichter
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany; Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Mila Marie Paul
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Martin Pauli
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
| | - Susanne Schoch
- Department of Neuropathology and Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius-Maximilians-University Würzburg, 97074 Würzburg, Germany
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Würzburg, 97074 Würzburg, Germany.
| | - Manfred Heckmann
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany.
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany; Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany.
| |
Collapse
|
15
|
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: 27] [Impact Index Per Article: 13.5] [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
|
16
|
Yu Z, Sakai M, Fukushima H, Ono C, Kikuchi Y, Koyama R, Matsui K, Furuyashiki T, Kida S, Tomita H. Contextual fear conditioning regulates synapse-related gene transcription in mouse microglia. Brain Res Bull 2022; 189:57-68. [PMID: 35987296 DOI: 10.1016/j.brainresbull.2022.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/04/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Microglia have been suggested to be involved in the underlying mechanism of conditional fear memory formation by regulating inflammatory cytokines. However, the mechanism linking microglia and neuronal activity related to fear conditioning remains unclear. This study characterized the transcription profile of microglia in a fear memory conditional mouse model. Compared with those in control mice microglia, the most significantly induced genes were synapse-related, whereas immune-related genes were reduced due to fear memory consolidation. Whilst the increased expression of synapse-related genes was reversed after fear memory extinction, that of immunological genes was not, strongly suggesting a connection between microglia, neurons, and a dysregulated immune response following contextual fear conditioning. Furthermore, in the hippocampal microglia, we found that the expression of neurotransmitter release regulators, γ-aminobutyric acid (GABA) receptor GABRB3 and synapsin 1/2, increased under fear memory consolidation and restored (decreased) after extinction. In addition, compared with the transcription profile in peripheral monocytes, few overlapping genes were not enriched in biological processes. Taken together, the identified conditional fear stress-induced changes in mouse microglial transcription profiles suggest that microglia-neuron communication mediates contextual fear conditioning.
Collapse
Affiliation(s)
- Zhiqian Yu
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.
| | - Mai Sakai
- Department of Psychiatry Nursing, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hotaka Fukushima
- Department of Bioscience, Faculty of Life Sciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Chiaki Ono
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yoshie Kikuchi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Satoshi Kida
- Department of Bioscience, Faculty of Life Sciences, Tokyo University of Agriculture, Tokyo, Japan; Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Disaster Psychiatry, International Research Institute for Disaster Science, Tohoku University, Sendai, Japan
| |
Collapse
|
17
|
Siller A, Hofer NT, Tomagra G, Burkert N, Hess S, Benkert J, Gaifullina A, Spaich D, Duda J, Poetschke C, Vilusic K, Fritz EM, Schneider T, Kloppenburg P, Liss B, Carabelli V, Carbone E, Ortner NJ, Striessnig J. β2-subunit alternative splicing stabilizes Cav2.3 Ca 2+ channel activity during continuous midbrain dopamine neuron-like activity. eLife 2022; 11:e67464. [PMID: 35792082 PMCID: PMC9307272 DOI: 10.7554/elife.67464] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
In dopaminergic (DA) Substantia nigra (SN) neurons Cav2.3 R-type Ca2+-currents contribute to somatodendritic Ca2+-oscillations. This activity may contribute to the selective degeneration of these neurons in Parkinson's disease (PD) since Cav2.3-knockout is neuroprotective in a PD mouse model. Here, we show that in tsA-201-cells the membrane-anchored β2-splice variants β2a and β2e are required to stabilize Cav2.3 gating properties allowing sustained Cav2.3 availability during simulated pacemaking and enhanced Ca2+-currents during bursts. We confirmed the expression of β2a- and β2e-subunit transcripts in the mouse SN and in identified SN DA neurons. Patch-clamp recordings of mouse DA midbrain neurons in culture and SN DA neurons in brain slices revealed SNX-482-sensitive R-type Ca2+-currents with voltage-dependent gating properties that suggest modulation by β2a- and/or β2e-subunits. Thus, β-subunit alternative splicing may prevent a fraction of Cav2.3 channels from inactivation in continuously active, highly vulnerable SN DA neurons, thereby also supporting Ca2+ signals contributing to the (patho)physiological role of Cav2.3 channels in PD.
Collapse
Affiliation(s)
- Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Giulia Tomagra
- Department of Drug Science, NIS Centre, University of TorinoTorinoItaly
| | - Nicole Burkert
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Simon Hess
- Institute for Zoology, Biocenter, University of CologneCologneGermany
| | - Julia Benkert
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Aisylu Gaifullina
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Desiree Spaich
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Johanna Duda
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | | | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Toni Schneider
- Institute of Neurophysiology, University of CologneCologneGermany
| | - Peter Kloppenburg
- Institute for Zoology, Biocenter, University of CologneCologneGermany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
- Linacre College & New College, University of OxfordOxfordUnited Kingdom
| | | | - Emilio Carbone
- Department of Drug Science, NIS Centre, University of TorinoTorinoItaly
| | - Nadine Jasmin Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| |
Collapse
|
18
|
Identification of ultra-rare disruptive variants in voltage-gated calcium channel-encoding genes in Japanese samples of schizophrenia and autism spectrum disorder. Transl Psychiatry 2022; 12:84. [PMID: 35220405 PMCID: PMC8882172 DOI: 10.1038/s41398-022-01851-y] [Citation(s) in RCA: 3] [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: 01/20/2022] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
Several large-scale whole-exome sequencing studies in patients with schizophrenia (SCZ) and autism spectrum disorder (ASD) have identified rare variants with modest or strong effect size as genetic risk factors. Dysregulation of cellular calcium homeostasis might be involved in SCZ/ASD pathogenesis, and genes encoding L-type voltage-gated calcium channel (VGCC) subunits Cav1.1 (CACNA1S), Cav1.2 (CACNA1C), Cav1.3 (CACNA1D), and T-type VGCC subunit Cav3.3 (CACNA1I) recently were identified as risk loci for psychiatric disorders. We performed a screening study, using the Ion Torrent Personal Genome Machine (PGM), of exon regions of these four candidate genes (CACNA1C, CACNA1D, CACNA1S, CACNA1I) in 370 Japanese patients with SCZ and 192 with ASD. Variant filtering was applied to identify biologically relevant mutations that were not registered in the dbSNP database or that have a minor allele frequency of less than 1% in East-Asian samples from databases; and are potentially disruptive, including nonsense, frameshift, canonical splicing site single nucleotide variants (SNVs), and non-synonymous SNVs predicted as damaging by five different in silico analyses. Each of these filtered mutations were confirmed by Sanger sequencing. If parental samples were available, segregation analysis was employed for measuring the inheritance pattern. Using our filter, we discovered one nonsense SNV (p.C1451* in CACNA1D), one de novo SNV (p.A36V in CACNA1C), one rare short deletion (p.E1675del in CACNA1D), and 14 NSstrict SNVs (non-synonymous SNV predicted as damaging by all of five in silico analyses). Neither p.A36V in CACNA1C nor p.C1451* in CACNA1D were found in 1871 SCZ cases, 380 ASD cases, or 1916 healthy controls in the independent sample set, suggesting that these SNVs might be ultra-rare SNVs in the Japanese population. The neuronal splicing isoform of Cav1.2 with the p.A36V mutation, discovered in the present study, showed reduced Ca2+-dependent inhibition, resulting in excessive Ca2+ entry through the mutant channel. These results suggested that this de novo SNV in CACNA1C might predispose to SCZ by affecting Ca2+ homeostasis. Thus, our analysis successfully identified several ultra-rare and potentially disruptive gene variants, lending partial support to the hypothesis that VGCC-encoding genes may contribute to the risk of SCZ/ASD.
Collapse
|
19
|
Takikawa K, Nishimune H. Similarity and Diversity of Presynaptic Molecules at Neuromuscular Junctions and Central Synapses. Biomolecules 2022; 12:biom12020179. [PMID: 35204679 PMCID: PMC8961632 DOI: 10.3390/biom12020179] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
Synaptic transmission is essential for controlling motor functions and maintaining brain functions such as walking, breathing, cognition, learning, and memory. Neurotransmitter release is regulated by presynaptic molecules assembled in active zones of presynaptic terminals. The size of presynaptic terminals varies, but the size of a single active zone and the types of presynaptic molecules are highly conserved among neuromuscular junctions (NMJs) and central synapses. Three parameters play an important role in the determination of neurotransmitter release properties at NMJs and central excitatory/inhibitory synapses: the number of presynaptic molecular clusters, the protein families of the presynaptic molecules, and the distance between presynaptic molecules and voltage-gated calcium channels. In addition, dysfunction of presynaptic molecules causes clinical symptoms such as motor and cognitive decline in patients with various neurological disorders and during aging. This review focuses on the molecular mechanisms responsible for the functional similarities and differences between excitatory and inhibitory synapses in the peripheral and central nervous systems, and summarizes recent findings regarding presynaptic molecules assembled in the active zone. Furthermore, we discuss the relationship between functional alterations of presynaptic molecules and dysfunction of NMJs or central synapses in diseases and during aging.
Collapse
Affiliation(s)
- Kenji Takikawa
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan;
| | - Hiroshi Nishimune
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan;
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan
- Correspondence: ; Tel.: +81-3-3964-3241
| |
Collapse
|
20
|
Paul MM, Dannhäuser S, Morris L, Mrestani A, Hübsch M, Gehring J, Hatzopoulos GN, Pauli M, Auger GM, Bornschein G, Scholz N, Ljaschenko D, Müller M, Sauer M, Schmidt H, Kittel RJ, DiAntonio A, Vakonakis I, Heckmann M, Langenhan T. The human cognition-enhancing CORD7 mutation increases active zone number and synaptic release. Brain 2022; 145:3787-3802. [DOI: 10.1093/brain/awac011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/29/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
Abstract
Humans carrying the CORD7 (cone-rod dystrophy 7) mutation possess increased verbal IQ and working memory. This autosomal dominant syndrome is caused by the single-amino acid R844H exchange (human numbering) located in the 310 helix of the C2A domain of RIMS1/RIM1 (Rab3-interacting molecule 1). RIM is an evolutionarily conserved multi-domain protein and essential component of presynaptic active zones, which is centrally involved in fast, Ca2+-triggered neurotransmitter release. How the CORD7 mutation affects synaptic function has remained unclear thus far. Here, we established Drosophila melanogaster as a disease model for clarifying the effects of the CORD7 mutation on RIM function and synaptic vesicle release.
To this end, using protein expression and X-ray crystallography, we solved the molecular structure of the Drosophila C2A domain at 1.92 Å resolution and by comparison to its mammalian homolog ascertained that the location of the CORD7 mutation is structurally conserved in fly RIM. Further, CRISPR/Cas9-assisted genomic engineering was employed for the generation of rim alleles encoding the R915H CORD7 exchange or R915E,R916E substitutions (fly numbering) to effect local charge reversal at the 310 helix. Through electrophysiological characterization by two-electrode voltage clamp and focal recordings we determined that the CORD7 mutation exerts a semi-dominant rather than a dominant effect on synaptic transmission resulting in faster, more efficient synaptic release and increased size of the readily releasable pool but decreased sensitivity for the fast calcium chelator BAPTA. In addition, the rim CORD7 allele increased the number of presynaptic active zones but left their nanoscopic organization unperturbed as revealed by super-resolution microscopy of the presynaptic scaffold protein Bruchpilot/ELKS/CAST.
We conclude that the CORD7 mutation leads to tighter release coupling, an increased readily releasable pool size and more release sites thereby promoting more efficient synaptic transmitter release. These results strongly suggest that similar mechanisms may underlie the CORD7 disease phenotype in patients and that enhanced synaptic transmission may contribute to their increased cognitive abilities.
Collapse
Affiliation(s)
- Mila M. Paul
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
- Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Lydia Morris
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Achmed Mrestani
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Martha Hübsch
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Jennifer Gehring
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | | | - Martin Pauli
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Genevieve M. Auger
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Grit Bornschein
- Carl Ludwig Institute of Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Nicole Scholz
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Dmitrij Ljaschenko
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Martin Müller
- Department of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, 97074 Würzburg, Germany
| | - Hartmut Schmidt
- Carl Ludwig Institute of Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Robert J. Kittel
- Carl Ludwig Institute of Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Department of Animal Physiology, Institute of Biology, Leipzig University, 04103 Leipzig, Germany
| | - Aaron DiAntonio
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | | | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Tobias Langenhan
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
21
|
Szule JA. Hypothesis Relating the Structure, Biochemistry and Function of Active Zone Material Macromolecules at a Neuromuscular Junction. Front Synaptic Neurosci 2022; 13:798225. [PMID: 35069169 PMCID: PMC8766674 DOI: 10.3389/fnsyn.2021.798225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022] Open
Abstract
This report integrates knowledge of in situ macromolecular structures and synaptic protein biochemistry to propose a unified hypothesis for the regulation of certain vesicle trafficking events (i.e., docking, priming, Ca2+-triggering, and membrane fusion) that lead to neurotransmitter secretion from specialized “active zones” of presynaptic axon terminals. Advancements in electron tomography, to image tissue sections in 3D at nanometer scale resolution, have led to structural characterizations of a network of different classes of macromolecules at the active zone, called “Active Zone Material’. At frog neuromuscular junctions, the classes of Active Zone Material macromolecules “top-masts”, “booms”, “spars”, “ribs” and “pins” direct synaptic vesicle docking while “pins”, “ribs” and “pegs” regulate priming to influence Ca2+-triggering and membrane fusion. Other classes, “beams”, “steps”, “masts”, and “synaptic vesicle luminal filaments’ likely help organize and maintain the structural integrity of active zones. Extensive studies on the biochemistry that regulates secretion have led to comprehensive characterizations of the many conserved proteins universally involved in these trafficking events. Here, a hypothesis including a partial proteomic atlas of Active Zone Material is presented which considers the common roles, binding partners, physical features/structure, and relative positioning in the axon terminal of both the proteins and classes of macromolecules involved in the vesicle trafficking events. The hypothesis designates voltage-gated Ca2+ channels and Ca2+-gated K+ channels to ribs and pegs that are connected to macromolecules that span the presynaptic membrane at the active zone. SNARE proteins (Syntaxin, SNAP25, and Synaptobrevin), SNARE-interacting proteins Synaptotagmin, Munc13, Munc18, Complexin, and NSF are designated to ribs and/or pins. Rab3A and Rabphillin-3A are designated to top-masts and/or booms and/or spars. RIM, Bassoon, and Piccolo are designated to beams, steps, masts, ribs, spars, booms, and top-masts. Spectrin is designated to beams. Lastly, the luminal portions of SV2 are thought to form the bulk of the observed synaptic vesicle luminal filaments. The goal here is to help direct future studies that aim to bridge Active Zone Material structure, biochemistry, and function to ultimately determine how it regulates the trafficking events in vivo that lead to neurotransmitter secretion.
Collapse
|
22
|
Hilton BJ, Husch A, Schaffran B, Lin TC, Burnside ER, Dupraz S, Schelski M, Kim J, Müller JA, Schoch S, Imig C, Brose N, Bradke F. An active vesicle priming machinery suppresses axon regeneration upon adult CNS injury. Neuron 2022; 110:51-69.e7. [PMID: 34706221 PMCID: PMC8730507 DOI: 10.1016/j.neuron.2021.10.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
Axons in the adult mammalian central nervous system fail to regenerate after spinal cord injury. Neurons lose their capacity to regenerate during development, but the intracellular processes underlying this loss are unclear. We found that critical components of the presynaptic active zone prevent axon regeneration in adult mice. Transcriptomic analysis combined with live-cell imaging revealed that adult primary sensory neurons downregulate molecular constituents of the synapse as they acquire the ability to rapidly grow their axons. Pharmacogenetic reduction of neuronal excitability stimulated axon regeneration after adult spinal cord injury. Genetic gain- and loss-of-function experiments uncovered that essential synaptic vesicle priming proteins of the presynaptic active zone, but not clostridial-toxin-sensitive VAMP-family SNARE proteins, inhibit axon regeneration. Systemic administration of Baclofen reduced voltage-dependent Ca2+ influx in primary sensory neurons and promoted their regeneration after spinal cord injury. These findings indicate that functional presynaptic active zones constitute a major barrier to axon regeneration.
Collapse
Affiliation(s)
- Brett J Hilton
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Andreas Husch
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Barbara Schaffran
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Tien-Chen Lin
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Emily R Burnside
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Sebastian Dupraz
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Max Schelski
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Jisoo Kim
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Susanne Schoch
- Institute of Neuropathology, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Cordelia Imig
- 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
| | - Frank Bradke
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
23
|
Piekut T, Wong YY, Walker SE, Smith CL, Gauberg J, Harracksingh AN, Lowden C, Novogradac BB, Cheng HYM, Spencer GE, Senatore A. Early Metazoan Origin and Multiple Losses of a Novel Clade of RIM Presynaptic Calcium Channel Scaffolding Protein Homologs. Genome Biol Evol 2021; 12:1217-1239. [PMID: 32413100 PMCID: PMC7456537 DOI: 10.1093/gbe/evaa097] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
The precise localization of CaV2 voltage-gated calcium channels at the synapse active zone requires various interacting proteins, of which, Rab3-interacting molecule or RIM is considered particularly important. In vertebrates, RIM interacts with CaV2 channels in vitro via a PDZ domain that binds to the extreme C-termini of the channels at acidic ligand motifs of D/E-D/E/H-WC-COOH, and knockout of RIM in vertebrates and invertebrates disrupts CaV2 channel synaptic localization and synapse function. Here, we describe a previously uncharacterized clade of RIM proteins bearing domain architectures homologous to those of known RIM homologs, but with some notable differences including key amino acids associated with PDZ domain ligand specificity. This novel RIM emerged near the stem lineage of metazoans and underwent extensive losses, but is retained in select animals including the early-diverging placozoan Trichoplax adhaerens, and molluscs. RNA expression and localization studies in Trichoplax and the mollusc snail Lymnaea stagnalis indicate differential regional/tissue type expression, but overlapping expression in single isolated neurons from Lymnaea. Ctenophores, the most early-diverging animals with synapses, are unique among animals with nervous systems in that they lack the canonical RIM, bearing only the newly identified homolog. Through phylogenetic analysis, we find that CaV2 channel D/E-D/E/H-WC-COOH like PDZ ligand motifs were present in the common ancestor of cnidarians and bilaterians, and delineate some deeply conserved C-terminal structures that distinguish CaV1 from CaV2 channels, and CaV1/CaV2 from CaV3 channels.
Collapse
Affiliation(s)
| | | | - Sarah E Walker
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Carolyn L Smith
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | | |
Collapse
|
24
|
Abstract
The chronification of pain can be attributed to changes in membrane receptors and channels underlying neuronal plasticity and signal transduction largely within nociceptive neurons that initiate and maintain pathological pain states. These proteins are subject to dynamic modification by posttranslational modifications, creating a code that controls protein function in time and space. Phosphorylation is an important posttranslational modification that affects ∼30% of proteins in vivo. Increased phosphorylation of various nociceptive ion channels and of their modulators underlies sensitization of different pain states. Cyclin-dependent kinases are proline-directed serine/threonine kinases that impact various biological and cellular systems. Cyclin-dependent kinase 5 (Cdk5), one member of this kinase family, and its activators p35 and p39 are expressed in spinal nerves, dorsal root ganglia, and the dorsal horn of the spinal cord. In neuropathic pain conditions, expression and/or activity of Cdk5 is increased, implicating Cdk5 in nociception. Experimental evidence suggests that Cdk5 is regulated through its own phosphorylation, through increasing p35's interaction with Cdk5, and through cleavage of p35 into p25. This narrative review discusses the molecular mechanisms of Cdk5-mediated regulation of target proteins involved in neuropathic pain. We focus on Cdk5 substrates that have been linked to nociceptive pathways, including channels (eg, transient receptor potential cation channel and voltage-gated calcium channel), proteins involved in neurotransmitter release (eg, synaptophysin and collapsin response mediator protein 2), and receptors (eg, glutamate, purinergic, and opioid). By altering the phosphoregulatory "set point" of proteins involved in pain signaling, Cdk5 thus appears to be an attractive target for treating neuropathic pain conditions.
Collapse
|
25
|
Young SM, Veeraraghavan P. Presynaptic voltage-gated calcium channels in the auditory brainstem. Mol Cell Neurosci 2021; 112:103609. [PMID: 33662542 DOI: 10.1016/j.mcn.2021.103609] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022] Open
Abstract
Sound information encoding within the initial synapses in the auditory brainstem requires reliable and precise synaptic transmission in response to rapid and large fluctuations in action potential (AP) firing rates. The magnitude and location of Ca2+ entry through voltage-gated Ca2+ channels (CaV) in the presynaptic terminal are key determinants in triggering AP-mediated release. In the mammalian central nervous system (CNS), the CaV2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing.
Collapse
Affiliation(s)
- Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | |
Collapse
|
26
|
Gandini MA, Zamponi GW. Voltage‐gated calcium channel nanodomains: molecular composition and function. FEBS J 2021; 289:614-633. [DOI: 10.1111/febs.15759] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Maria A. Gandini
- Department of Physiology and Pharmacology Alberta Children’s Hospital Research Institute Hotchkiss Brain Institute Cumming School of Medicine University of Calgary AB Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology Alberta Children’s Hospital Research Institute Hotchkiss Brain Institute Cumming School of Medicine University of Calgary AB Canada
| |
Collapse
|
27
|
Gauberg J, Abdallah S, Elkhatib W, Harracksingh AN, Piekut T, Stanley EF, Senatore A. Conserved biophysical features of the Ca V2 presynaptic Ca 2+ channel homologue from the early-diverging animal Trichoplax adhaerens. J Biol Chem 2020; 295:18553-18578. [PMID: 33097592 PMCID: PMC7939481 DOI: 10.1074/jbc.ra120.015725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/21/2020] [Indexed: 12/20/2022] Open
Abstract
The dominant role of CaV2 voltage-gated calcium channels for driving neurotransmitter release is broadly conserved. Given the overlapping functional properties of CaV2 and CaV1 channels, and less so CaV3 channels, it is unclear why there have not been major shifts toward dependence on other CaV channels for synaptic transmission. Here, we provide a structural and functional profile of the CaV2 channel cloned from the early-diverging animal Trichoplax adhaerens, which lacks a nervous system but possesses single gene homologues for CaV1-CaV3 channels. Remarkably, the highly divergent channel possesses similar features as human CaV2.1 and other CaV2 channels, including high voltage-activated currents that are larger in external Ba2+ than in Ca2+; voltage-dependent kinetics of activation, inactivation, and deactivation; and bimodal recovery from inactivation. Altogether, the functional profile of Trichoplax CaV2 suggests that the core features of presynaptic CaV2 channels were established early during animal evolution, after CaV1 and CaV2 channels emerged via proposed gene duplication from an ancestral CaV1/2 type channel. The Trichoplax channel was relatively insensitive to mammalian CaV2 channel blockers ω-agatoxin-IVA and ω-conotoxin-GVIA and to metal cation blockers Cd2+ and Ni2+ Also absent was the capacity for voltage-dependent G-protein inhibition by co-expressed Trichoplax Gβγ subunits, which nevertheless inhibited the human CaV2.1 channel, suggesting that this modulatory capacity evolved via changes in channel sequence/structure, and not G proteins. Last, the Trichoplax channel was immunolocalized in cells that express an endomorphin-like peptide implicated in cell signaling and locomotive behavior and other likely secretory cells, suggesting contributions to regulated exocytosis.
Collapse
Affiliation(s)
- Julia Gauberg
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Salsabil Abdallah
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Wassim Elkhatib
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Alicia N Harracksingh
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Thomas Piekut
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Elise F Stanley
- Laboratory of Synaptic Transmission, Krembil Research Institute, Toronto, Ontario, Canada
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| |
Collapse
|
28
|
Disentangling the Roles of RIM and Munc13 in Synaptic Vesicle Localization and Neurotransmission. J Neurosci 2020; 40:9372-9385. [PMID: 33139401 PMCID: PMC7724145 DOI: 10.1523/jneurosci.1922-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/22/2020] [Accepted: 10/15/2020] [Indexed: 11/21/2022] Open
Abstract
Efficient neurotransmitter release at the presynaptic terminal requires docking of synaptic vesicles to the active zone membrane and formation of fusion-competent synaptic vesicles near voltage-gated Ca2+ channels. Rab3-interacting molecule (RIM) is a critical active zone organizer, as it recruits Ca2+ channels and activates synaptic vesicle docking and priming via Munc13-1. However, our knowledge about Munc13-independent contributions of RIM to active zone functions is limited. To identify the functions that are solely mediated by RIM, we used genetic manipulations to control RIM and Munc13-1 activity in cultured hippocampal neurons from mice of either sex and compared synaptic ultrastructure and neurotransmission. We found that RIM modulates synaptic vesicle localization in the proximity of the active zone membrane independent of Munc13-1. In another step, both RIM and Munc13 mediate synaptic vesicle docking and priming. In addition, while the activity of both RIM and Munc13-1 is required for Ca2+-evoked release, RIM uniquely controls neurotransmitter release efficiency. However, activity-dependent augmentation of synaptic vesicle pool size relies exclusively on the action of Munc13s. Based on our results, we extend previous findings and propose a refined model in which RIM and Munc13-1 act in overlapping and independent stages of synaptic vesicle localization and release. SIGNIFICANCE STATEMENT The presynaptic active zone is composed of scaffolding proteins that functionally interact to localize synaptic vesicles to release sites, ensuring neurotransmission. Our current knowledge of the presynaptic active zone function relies on structure-function analysis, which has provided detailed information on the network of interactions and the impact of active zone proteins. Yet, the hierarchical, redundant, or independent cooperation of each active zone protein to synapse functions is not fully understood. Rab3-interacting molecule and Munc13 are the two key functionally interacting active zone proteins. Here, we dissected the distinct actions of Rab3-interacting molecule and Munc13-1 from both ultrastructural and physiological aspects. Our findings provide a more detailed view of how these two presynaptic proteins orchestrate their functions to achieve synaptic transmission.
Collapse
|
29
|
Sameera, Shah FA, Rashid S. Conformational ensembles of non-peptide ω-conotoxin mimetics and Ca +2 ion binding to human voltage-gated N-type calcium channel Ca v2.2. Comput Struct Biotechnol J 2020; 18:2357-2372. [PMID: 32994894 PMCID: PMC7498737 DOI: 10.1016/j.csbj.2020.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic neuropathic pain is the most complex and challenging clinical problem of a population that sets a major physical and economic burden at the global level. Ca2+-permeable channels functionally orchestrate the processing of pain signals. Among them, N-type voltage-gated calcium channels (VGCC) hold prominent contribution in the pain signal transduction and serve as prime targets for synaptic transmission block and attenuation of neuropathic pain. Here, we present detailed in silico analysis to comprehend the underlying conformational changes upon Ca2+ ion passage through Cav2.2 to differentially correlate subtle transitions induced via binding of a conopeptide-mimetic alkylphenyl ether-based analogue MVIIA. Interestingly, pronounced conformational changes were witnessed at the proximal carboxyl-terminus of Cav2.2 that attained an upright orientation upon Ca+2 ion permeability. Moreover, remarkable changes were observed in the architecture of channel tunnel. These findings illustrate that inhibitor binding to Cav2.2 may induce more narrowing in the pore size as compared to Ca2+ binding through modulating the hydrophilicity pattern at the selectivity region. A significant reduction in the tunnel volume at the selectivity filter and its enhancement at the activation gate of Ca+2-bound Cav2.2 suggests that ion binding modulates the outward splaying of pore-lining S6 helices to open the voltage gate. Overall, current study delineates dynamic conformational ensembles in terms of Ca+2 ion and MVIIA-associated structural implications in the Cav2.2 that may help in better therapeutic intervention to chronic and neuropathic pain management.
Collapse
Affiliation(s)
- Sameera
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
30
|
Abstract
Background Insulin is stored within large dense-core granules in pancreatic beta (β)-cells and is released by Ca2+-triggered exocytosis with increasing blood glucose levels. Polarized and targeted secretion of insulin from β-cells in pancreatic islets into the vasculature has been proposed; however, the mechanisms related to cellular and molecular localization remain largely unknown. Within nerve terminals, the Ca2+-dependent release of a polarized transmitter is limited to the active zone, a highly specialized area of the presynaptic membrane. Several active zone-specific proteins have been characterized; among them, the CAST/ELKS protein family members have the ability to form large protein complexes with other active zone proteins to control the structure and function of the active zone for tight regulation of neurotransmitter release. Notably, ELKS but not CAST is also expressed in β-cells, implying that ELKS may be involved in polarized insulin secretion from β-cells. Scope of review This review provides an overview of the current findings regarding the role(s) of ELKS and other active zone proteins in β-cells and focuses on the molecular mechanism underlying ELKS regulation within polarized insulin secretion from islets. Major conclusions ELKS localizes at the vascular-facing plasma membrane of β-cells in mouse pancreatic islets. ELKS forms a potent insulin secretion complex with L-type voltage-dependent Ca2+ channels on the vascular-facing plasma membrane of β-cells, enabling polarized Ca2+ influx and first-phase insulin secretion from islets. This model provides novel insights into the functional polarity observed during insulin secretion from β-cells within islets at the molecular level. This active zone-like region formed by ELKS at the vascular side of the plasma membrane is essential for coordinating physiological insulin secretion and may be disrupted in diabetes.
Collapse
Affiliation(s)
- Mica Ohara-Imaizumi
- Department of Cellular Biochemistry, Kyorin University School of Medicine, Tokyo 181-8611, Japan.
| | - Kyota Aoyagi
- Department of Cellular Biochemistry, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
31
|
Held RG, Liu C, Ma K, Ramsey AM, Tarr TB, De Nola G, Wang SSH, Wang J, van den Maagdenberg AMJM, Schneider T, Sun J, Blanpied TA, Kaeser PS. Synapse and Active Zone Assembly in the Absence of Presynaptic Ca 2+ Channels and Ca 2+ Entry. Neuron 2020; 107:667-683.e9. [PMID: 32616470 DOI: 10.1016/j.neuron.2020.05.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/24/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Presynaptic CaV2 channels are essential for Ca2+-triggered exocytosis. In addition, there are two competing models for their roles in synapse structure. First, Ca2+ channels or Ca2+ entry may control synapse assembly. Second, active zone proteins may scaffold CaV2s to presynaptic release sites, and synapse structure is CaV2 independent. Here, we ablated all three CaV2s using conditional knockout in cultured hippocampal neurons or at the calyx of Held, which abolished evoked exocytosis. Compellingly, synapse and active zone structure, vesicle docking, and transsynaptic nano-organization were unimpaired. Similarly, long-term blockade of action potentials and Ca2+ entry did not disrupt active zone assembly. Although CaV2 knockout impaired the localization of β subunits, α2δ-1 localized normally. Rescue with CaV2 restored exocytosis, and CaV2 active zone targeting depended on the intracellular C-terminus. We conclude that synapse assembly is independent of CaV2s or Ca2+ entry through them. Instead, active zone proteins recruit and anchor CaV2s via CaV2 C-termini.
Collapse
Affiliation(s)
- Richard G Held
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kunpeng Ma
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 100101, China
| | - Austin M Ramsey
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tyler B Tarr
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Giovanni De Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jiexin Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Toni Schneider
- Institute for Neurophysiology, University of Cologne, Köln 50931, Germany
| | - Jianyuan Sun
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 100101, China
| | - Thomas A Blanpied
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Identification of Novel Targets of RBM5 in the Healthy and Injured Brain. Neuroscience 2020; 440:299-315. [PMID: 32335213 DOI: 10.1016/j.neuroscience.2020.04.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 11/20/2022]
Abstract
The tumor suppressor RNA-binding motif 5 (RBM5) regulates the expression levels and cassette exon-definition (i.e. splicing) of a select set of mRNAs in a tissue-specific manner. Most RBM5-regulated targets were identified in oncological investigations and frequently involve genes which mediate apoptotic cell death. Little is known about the role of RBM5 in the brain. Also, it is unclear if a brain injury may be required to detect RBM5 mediated effects on pro-apoptotic genes due to their low expression levels in the healthy adult CNS at baseline. Conditional/floxed (brain-specific) gene deleter mice were generated to elucidate CNS-specific RBM5 mRNA targets. Male/female mice were subjected to a severe controlled cortical impact (CCI) traumatic brain injury (TBI) in order to increase the background expression of pro-death mRNAs and facilitate testing of the hypothesis that RBM5 inhibition decreases post-injury upregulation of caspases/FAS in the CNS. As expected, a CCI increased caspases/FAS mRNA in the injured cortex. RBM5 KO did not affect their levels or splicing. Surprisingly, KO increased the mRNA levels of novel targets including casein kinase 2 alpha prime interacting protein (Csnka2ip/CKT2) - a gene not thought to be expressed in the brain, contrary to findings here. Twenty-two unique splicing events were also detected in KOs including increased block-inclusion of cassette exons 20-22 in regulating synaptic membrane exocytosis 2 (Rims2). In conclusion, here we used genome-wide transcriptomic analysis on healthy and injured RBM5 KO mouse brain tissue to elucidate the first known gene targets of this enigmatic RBP in this CNS.
Collapse
|
33
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
34
|
ELKS/Voltage-Dependent Ca 2+ Channel-β Subunit Module Regulates Polarized Ca 2+ Influx in Pancreatic β Cells. Cell Rep 2020; 26:1213-1226.e7. [PMID: 30699350 DOI: 10.1016/j.celrep.2018.12.106] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 10/29/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
Pancreatic β cells secrete insulin by Ca2+-triggered exocytosis. However, there is no apparent secretory site similar to the neuronal active zones, and the cellular and molecular localization mechanism underlying polarized exocytosis remains elusive. Here, we report that ELKS, a vertebrate active zone protein, is used in β cells to regulate Ca2+ influx for insulin secretion. β cell-specific ELKS-knockout (KO) mice showed impaired glucose-stimulated first-phase insulin secretion and reduced L-type voltage-dependent Ca2+ channel (VDCC) current density. In situ Ca2+ imaging of β cells within islets expressing a membrane-bound G-CaMP8b Ca2+ sensor demonstrated initial local Ca2+ signals at the ELKS-localized vascular side of the β cell plasma membrane, which were markedly decreased in ELKS-KO β cells. Mechanistically, ELKS directly interacted with the VDCC-β subunit via the GK domain. These findings suggest that ELKS and VDCCs form a potent insulin secretion complex at the vascular side of the β cell plasma membrane for polarized Ca2+ influx and first-phase insulin secretion from pancreatic islets.
Collapse
|
35
|
Autism-associated mutations in the CaVβ2 calcium-channel subunit increase Ba2+-currents and lead to differential modulation by the RGK-protein Gem. Neurobiol Dis 2020; 136:104721. [DOI: 10.1016/j.nbd.2019.104721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/06/2019] [Accepted: 12/26/2019] [Indexed: 12/26/2022] Open
|
36
|
Moser T, Grabner CP, Schmitz F. Sensory Processing at Ribbon Synapses in the Retina and the Cochlea. Physiol Rev 2020; 100:103-144. [DOI: 10.1152/physrev.00026.2018] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent years, sensory neuroscientists have made major efforts to dissect the structure and function of ribbon synapses which process sensory information in the eye and ear. This review aims to summarize our current understanding of two key aspects of ribbon synapses: 1) their mechanisms of exocytosis and endocytosis and 2) their molecular anatomy and physiology. Our comparison of ribbon synapses in the cochlea and the retina reveals convergent signaling mechanisms, as well as divergent strategies in different sensory systems.
Collapse
Affiliation(s)
- Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| | - Chad P. Grabner
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| | - Frank Schmitz
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| |
Collapse
|
37
|
Ortner NJ, Pinggera A, Hofer NT, Siller A, Brandt N, Raffeiner A, Vilusic K, Lang I, Blum K, Obermair GJ, Stefan E, Engel J, Striessnig J. RBP2 stabilizes slow Cav1.3 Ca 2+ channel inactivation properties of cochlear inner hair cells. Pflugers Arch 2019; 472:3-25. [PMID: 31848688 PMCID: PMC6960213 DOI: 10.1007/s00424-019-02338-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 12/04/2019] [Indexed: 01/31/2023]
Abstract
Cav1.3 L-type Ca2+ channels (LTCCs) in cochlear inner hair cells (IHCs) are essential for hearing as they convert sound-induced graded receptor potentials into tonic postsynaptic glutamate release. To enable fast and indefatigable presynaptic Ca2+ signaling, IHC Cav1.3 channels exhibit a negative activation voltage range and uniquely slow inactivation kinetics. Interaction with CaM-like Ca2+-binding proteins inhibits Ca2+-dependent inactivation, while the mechanisms underlying slow voltage-dependent inactivation (VDI) are not completely understood. Here we studied if the complex formation of Cav1.3 LTCCs with the presynaptic active zone proteins RIM2α and RIM-binding protein 2 (RBP2) can stabilize slow VDI. We detected both RIM2α and RBP isoforms in adult mouse IHCs, where they co-localized with Cav1.3 and synaptic ribbons. Using whole-cell patch-clamp recordings (tsA-201 cells), we assessed their effect on the VDI of the C-terminal full-length Cav1.3 (Cav1.3L) and a short splice variant (Cav1.342A) that lacks the C-terminal RBP2 interaction site. When co-expressed with the auxiliary β3 subunit, RIM2α alone (Cav1.342A) or RIM2α/RBP2 (Cav1.3L) reduced Cav1.3 VDI to a similar extent as observed in IHCs. Membrane-anchored β2 variants (β2a, β2e) that inhibit inactivation on their own allowed no further modulation of inactivation kinetics by RIM2α/RBP2. Moreover, association with RIM2α and/or RBP2 consolidated the negative Cav1.3 voltage operating range by shifting the channel's activation threshold toward more hyperpolarized potentials. Taken together, the association with "slow" β subunits (β2a, β2e) or presynaptic scaffolding proteins such as RIM2α and RBP2 stabilizes physiological gating properties of IHC Cav1.3 LTCCs in a splice variant-dependent manner ensuring proper IHC function.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Niels Brandt
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Andrea Raffeiner
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Isabelle Lang
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Gerald J Obermair
- Division of Physiology, Medical University Innsbruck, Innsbruck, Austria.,Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Eduard Stefan
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Jutta Engel
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
38
|
Gandini MA, Souza IA, Raval D, Xu J, Pan YX, Zamponi GW. Differential regulation of Cav2.2 channel exon 37 variants by alternatively spliced μ-opioid receptors. Mol Brain 2019; 12:98. [PMID: 31775826 PMCID: PMC6880636 DOI: 10.1186/s13041-019-0524-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022] Open
Abstract
We have examined the regulation of mutually exclusive Cav2.2 exon 37a and b variants by the mouse μ-opioid receptor (mMOR) C-terminal splice variants 1, 1C and 1O in tsA-201 cells. Electrophysiological analyses revealed that both channel isoforms exhibit DAMGO-induced voltage-dependent (Gβγ-mediated) inhibition and its recovery by voltage pre-pulses, as well as a voltage-independent component. However, the two channel isoforms differ in their relative extent of voltage-dependent and independent inhibition, with Cav2.2-37b showing significantly more voltage-dependent inhibition upon activation of the three mMOR receptors studied. In addition, coexpression of either mMOR1 or mMOR1C results in an agonist-independent reduction in the peak current density of Cav2.2-37a channels, whereas the peak current density of Cav2.2-37b does not appear to be affected. Interestingly, this decrease is not due to an effect on channel expression at the plasma membrane, as demonstrated by biotinylation experiments. We further examined the mechanism underlying the agonist-independent modulation of Cav2.2-37a by mMOR1C. Incubation of cells with pertussis toxin did not affect the mMOR1C mediated inhibition of Cav2.2-37a currents, indicating a lack of involvement of Gi/o signaling. However, when a Src tyrosine kinase inhibitor was applied, the effect of mMOR1C was lost. Moreover, when we recorded currents using a Cav2.2-37a mutant in which tyrosine 1747 was replaced with phenylalanine (Y1747F), the agonist independent effects of mMOR1C were abolished. Altogether our findings show that Cav2.2-37a and Cav2.2-37b isoforms are subject to differential regulation by C-terminal splice variants of mMORs, and that constitutive mMOR1C activity and downstream tyrosine kinase activity exert a selective inhibition of the Cav2.2-37a splice variant, an N-type channel isoform that is highly enriched in nociceptors. Our study provides new insights into the roles of the MOR full-length C-terminal variants in modulating Cav2.2 channel isoform activities.
Collapse
Affiliation(s)
- Maria A Gandini
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ivana A Souza
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dvij Raval
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jin Xu
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ying-Xian Pan
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
39
|
Nanou E, Catterall WA. Calcium Channels, Synaptic Plasticity, and Neuropsychiatric Disease. Neuron 2019; 98:466-481. [PMID: 29723500 DOI: 10.1016/j.neuron.2018.03.017] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/06/2018] [Accepted: 03/09/2018] [Indexed: 12/14/2022]
Abstract
Voltage-gated calcium channels couple depolarization of the cell-surface membrane to entry of calcium, which triggers secretion, contraction, neurotransmission, gene expression, and other physiological responses. They are encoded by ten genes, which generate three voltage-gated calcium channel subfamilies: CaV1; CaV2; and CaV3. At synapses, CaV2 channels form large signaling complexes in the presynaptic nerve terminal, which are responsible for the calcium entry that triggers neurotransmitter release and short-term presynaptic plasticity. CaV1 channels form signaling complexes in postsynaptic dendrites and dendritic spines, where their calcium entry induces long-term potentiation. These calcium channels are the targets of mutations and polymorphisms that alter their function and/or regulation and cause neuropsychiatric diseases, including migraine headache, cerebellar ataxia, autism, schizophrenia, bipolar disorder, and depression. This article reviews the molecular properties of calcium channels, considers their multiple roles in synaptic plasticity, and discusses their potential involvement in this wide range of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Evanthia Nanou
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA.
| |
Collapse
|
40
|
Presynaptic Calcium Channels. Int J Mol Sci 2019; 20:ijms20092217. [PMID: 31064106 PMCID: PMC6539076 DOI: 10.3390/ijms20092217] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 12/27/2022] Open
Abstract
Presynaptic Ca2+ entry occurs through voltage-gated Ca2+ (CaV) channels which are activated by membrane depolarization. Depolarization accompanies neuronal firing and elevation of Ca2+ triggers neurotransmitter release from synaptic vesicles. For synchronization of efficient neurotransmitter release, synaptic vesicles are targeted by presynaptic Ca2+ channels forming a large signaling complex in the active zone. The presynaptic CaV2 channel gene family (comprising CaV2.1, CaV2.2, and CaV2.3 isoforms) encode the pore-forming α1 subunit. The cytoplasmic regions are responsible for channel modulation by interacting with regulatory proteins. This article overviews modulation of the activity of CaV2.1 and CaV2.2 channels in the control of synaptic strength and presynaptic plasticity.
Collapse
|
41
|
Guzman GA, Guzman RE, Jordan N, Hidalgo P. A Tripartite Interaction Among the Calcium Channel α 1- and β-Subunits and F-Actin Increases the Readily Releasable Pool of Vesicles and Its Recovery After Depletion. Front Cell Neurosci 2019; 13:125. [PMID: 31130843 PMCID: PMC6509170 DOI: 10.3389/fncel.2019.00125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/13/2019] [Indexed: 11/13/2022] Open
Abstract
Neurotransmitter release is initiated by the influx of Ca2+via voltage-gated calcium channels. The accessory β-subunit (CaVβ) of these channels shapes synaptic transmission by associating with the pore-forming subunit (CaVα1) and up-regulating presynaptic calcium currents. Besides CaVα1, CaVβ interacts with several partners including actin filaments (F-actin). These filaments are known to associate with synaptic vesicles (SVs) at the presynaptic terminals and support their translocation within different pools, but the role of CaVβ/F-actin association on synaptic transmission has not yet been explored. We here study how CaVβ4, the major calcium channel β isoform in mamalian brain, modifies synaptic transmission in concert with F-actin in cultured hippocampal neurons. We analyzed the effect of exogenous CaVβ4 before and after pharmacological disruption of the actin cytoskeleton and dissected calcium channel-dependent and -independent functions by comparing the effects of the wild-type subunit with the one bearing a double mutation that impairs binding to CaVα1. We found that exogenously expressed wild-type CaVβ4 enhances spontaneous and depolarization-evoked excitatory postsynaptic currents (EPSCs) without altering synaptogenesis. CaVβ4 increases the size of the readily releasable pool (RRP) of SVs at resting conditions and accelerates their recovery after depletion. The enhanced neurotransmitter release induced by CaVβ4 is abolished upon disruption of the actin cytoskeleton. The CaVα1 association-deficient CaVβ4 mutant associates with actin filaments, but neither alters postsynaptic responses nor the time course of the RRP recovery. Furthermore, this mutant protein preserves the ability to increase the RRP size. These results indicate that the interplay between CaVβ4 and F-actin also support the recruitment of SVs to the RRP in a CaVα1-independent manner. Our studies show an emerging role of CaVβ in determining SV maturation toward the priming state and its replenishment after release. We envision that this subunit plays a role in coupling exocytosis to endocytosis during the vesicle cycle.
Collapse
Affiliation(s)
- Gustavo A Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Raul E Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Nadine Jordan
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Patricia Hidalgo
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany.,Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
42
|
Heine M, Heck J, Ciuraszkiewicz A, Bikbaev A. Dynamic compartmentalization of calcium channel signalling in neurons. Neuropharmacology 2019; 169:107556. [PMID: 30851307 DOI: 10.1016/j.neuropharm.2019.02.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/16/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022]
Abstract
Calcium fluxes through the neuronal membrane are strictly limited in time due to biophysical properties of voltage-gated and ligand-activated ion channels and receptors. Being embedded into the crowded dynamic environment of biological membranes, Ca2+-permeable receptors and channels undergo perpetual spatial rearrangement, which enables their temporary association and formation of transient signalling complexes. Thus, efficient calcium-mediated signal transduction requires mechanisms to support very precise spatiotemporal alignment of the calcium source and Ca2+-binding lipids and proteins in a highly dynamic environment. The mobility of calcium channels and calcium-sensing proteins themselves can be considered as a physiologically meaningful variable that affects calcium-mediated signalling in neurons. In this review, we will focus on voltage-gated calcium channels (VGCCs) and activity-induced relocation of stromal interaction molecules (STIMs) in the endoplasmic reticulum (ER) to show that particularly in time ranges between milliseconds to minutes, dynamic rearrangement of calcium conducting channels and sensor molecules is of physiological relevance. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Martin Heine
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, 39106, Germany; RG Functional Neurobiology, Institute for Development Biology and Neurobiology, Johannes Gutenberg University Mainz, Germany.
| | - Jennifer Heck
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany
| | - Anna Ciuraszkiewicz
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, 39106, Germany
| | - Arthur Bikbaev
- RG Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, 39118, Germany
| |
Collapse
|
43
|
Pangrsic T, Singer JH, Koschak A. Voltage-Gated Calcium Channels: Key Players in Sensory Coding in the Retina and the Inner Ear. Physiol Rev 2019; 98:2063-2096. [PMID: 30067155 DOI: 10.1152/physrev.00030.2017] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Calcium influx through voltage-gated Ca (CaV) channels is the first step in synaptic transmission. This review concerns CaV channels at ribbon synapses in primary sense organs and their specialization for efficient coding of stimuli in the physical environment. Specifically, we describe molecular, biochemical, and biophysical properties of the CaV channels in sensory receptor cells of the retina, cochlea, and vestibular apparatus, and we consider how such properties might change over the course of development and contribute to synaptic plasticity. We pay particular attention to factors affecting the spatial arrangement of CaV channels at presynaptic, ribbon-type active zones, because the spatial relationship between CaV channels and release sites has been shown to affect synapse function critically in a number of systems. Finally, we review identified synaptopathies affecting sensory systems and arising from dysfunction of L-type, CaV1.3, and CaV1.4 channels or their protein modulatory elements.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Joshua H Singer
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Alexandra Koschak
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
44
|
Active Zone Proteins RIM1αβ Are Required for Normal Corticostriatal Transmission and Action Control. J Neurosci 2018; 39:1457-1470. [PMID: 30559150 DOI: 10.1523/jneurosci.1940-18.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/13/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
Dynamic regulation of synaptic transmission at cortical inputs to the dorsal striatum is considered critical for flexible and efficient action learning and control. Presynaptic mechanisms governing the properties and plasticity of glutamate release from these inputs are not fully understood, and the corticostriatal synaptic processes that support normal action learning and control remain unclear. Here we show in male and female mice that conditional deletion of presynaptic proteins RIM1αβ (RIM1) from excitatory cortical neurons impairs corticostriatal synaptic transmission in the dorsolateral striatum. Key forms of presynaptic G-protein-coupled receptor-mediated short- and long-term striatal plasticity are spared following RIM1 deletion. Conditional RIM1 KO mice show heightened novelty-induced locomotion and impaired motor learning on the accelerating rotarod. They further show heightened self-paced instrumental responding for food and impaired learning of a habitual instrumental response strategy. Together, these findings reveal a selective role for presynaptic RIM1 in neurotransmitter release at prominent basal ganglia synapses, and provide evidence that RIM1-dependent processes help to promote the refinement of skilled actions, constrain goal-directed behaviors, and support the learning and use of habits.SIGNIFICANCE STATEMENT Our daily functioning hinges on the ability to flexibly and efficiently learn and control our actions. How the brain encodes these capacities is unclear. Here we identified a selective role for presynaptic proteins RIM1αβ in controlling glutamate release from cortical inputs to the dorsolateral striatum, a brain structure critical for action learning and control. Behavioral analysis of mice with restricted genetic deletion of RIM1αβ further revealed roles for RIM1αβ-dependent processes in the learning and refinement of motor skills and the balanced expression of goal-directed and habitual actions.
Collapse
|
45
|
Hagiwara A, Kitahara Y, Grabner CP, Vogl C, Abe M, Kitta R, Ohta K, Nakamura K, Sakimura K, Moser T, Nishi A, Ohtsuka T. Cytomatrix proteins CAST and ELKS regulate retinal photoreceptor development and maintenance. J Cell Biol 2018; 217:3993-4006. [PMID: 30190286 PMCID: PMC6219712 DOI: 10.1083/jcb.201704076] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/18/2018] [Accepted: 08/03/2018] [Indexed: 01/06/2023] Open
Abstract
The retinal ribbon synapse is important for the processing of visual information. Hagiwara et al. show that the active zone proteins CAST and ELKS perform both redundant and unique functions in photoreceptors to promote the maturation, maintenance, and activity of ribbon synapses. At the presynaptic active zone (AZ), the related cytomatrix proteins CAST and ELKS organize the presynaptic release machinery. While CAST is known to regulate AZ size and neurotransmitter release, the role of ELKS and the integral system of CAST/ELKS together is poorly understood. Here, we show that CAST and ELKS have both redundant and unique roles in coordinating synaptic development, function, and maintenance of retinal photoreceptor ribbon synapses. A CAST/ELKS double knockout (dKO) mouse showed high levels of ectopic synapses and reduced responses to visual stimulation. Ectopic formation was not observed in ELKS conditional KO but progressively increased with age in CAST KO mice with higher rates in the dKO. Presynaptic calcium influx was strongly reduced in rod photoreceptors of CAST KO and dKO mice. Three-dimensional scanning EM reconstructions showed structural abnormalities in rod triads of CAST KO and dKO. Remarkably, AAV-mediated acute ELKS deletion after synapse maturation induced neurodegeneration and loss of ribbon synapses. These results suggest that CAST and ELKS work in concert to promote retinal synapse formation, transmission, and maintenance.
Collapse
Affiliation(s)
- Akari Hagiwara
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yosuke Kitahara
- Department of Pharmacology, Kurume University School of Medicine, Fukuoka, Japan
| | - Chad Paul Grabner
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christian Vogl
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Ryo Kitta
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keisuke Ohta
- Department of Anatomy, Kurume University School of Medicine, Fukuoka, Japan
| | - Keiichiro Nakamura
- Department of Anatomy, Kurume University School of Medicine, Fukuoka, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tobias Moser
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany .,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Fukuoka, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
46
|
Postsynaptic RIM1 modulates synaptic function by facilitating membrane delivery of recycling NMDARs in hippocampal neurons. Nat Commun 2018; 9:2267. [PMID: 29891949 PMCID: PMC5995852 DOI: 10.1038/s41467-018-04672-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/11/2018] [Indexed: 11/26/2022] Open
Abstract
NMDA receptors (NMDARs) are crucial for excitatory synaptic transmission and synaptic plasticity. The number and subunit composition of synaptic NMDARs are tightly controlled by neuronal activity and sensory experience, but the molecular mechanism mediating NMDAR trafficking remains poorly understood. Here, we report that RIM1, with a well-established role in presynaptic vesicle release, also localizes postsynaptically in the mouse hippocampus. Postsynaptic RIM1 in hippocampal CA1 region is required for basal NMDAR-, but not AMPA receptor (AMPAR)-, mediated synaptic responses, and contributes to synaptic plasticity and hippocampus-dependent memory. Moreover, RIM1 levels in hippocampal neurons influence both the constitutive and regulated NMDAR trafficking, without affecting constitutive AMPAR trafficking. We further demonstrate that RIM1 binds to Rab11 via its N terminus, and knockdown of RIM1 impairs membrane insertion of Rab11-positive recycling endosomes containing NMDARs. Together, these results identify a RIM1-dependent mechanism critical for modulating synaptic function by facilitating membrane delivery of recycling NMDARs. Rab3-interacting molecules (RIMs) are a key component of the presynaptic active zone that regulate neurotransmitter release. Here, the authors show that RIM1 also has postsynaptic function to organize NMDA receptors and synaptic response.
Collapse
|
47
|
Snidal CA, Li Q, Elliott BB, Mah HKH, Chen RHC, Gardezi SR, Stanley EF. Molecular Characterization of an SV Capture Site in the Mid-Region of the Presynaptic CaV2.1 Calcium Channel C-Terminal. Front Cell Neurosci 2018; 12:127. [PMID: 29867360 PMCID: PMC5958201 DOI: 10.3389/fncel.2018.00127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 04/23/2018] [Indexed: 01/28/2023] Open
Abstract
Neurotransmitter is released from presynaptic nerve terminals at fast-transmitting synapses by the action potential-gating of voltage dependent calcium channels (CaV), primarily of the CaV2.1 and CaV2.2 types. Entering Ca2+ diffuses to a nearby calcium sensor associated with a docked synaptic vesicle (SV) and initiates its fusion and discharge. Our previous findings that single CaVs can gate SV fusion argued for one or more tethers linking CaVs to docked SVs but the molecular nature of these tethers have not been established. We recently developed a cell-free, in vitro biochemical assay, termed SV pull-down (SV-PD), to test for SV binding proteins and used this to demonstrate that CaV2.2 or the distal third of its C-terminal can capture SVs. In subsequent reports we identified the binding site and characterized an SV binding motif. In this study, we set out to test if a similar SV-binding mechanism exists in the primary presynaptic channel type, CaV2.1. We cloned the chick variant of this channel and to our surprise found that it lacked the terminal third of the C-terminal, ruling out direct correlation with CaV2.2. We used SV-PD to identify an SV binding site in the distal half of the CaV2.1 C-terminal, a region that corresponds to the central third of the CaV2.2 C-terminal. Mutant fusion proteins combined with motif-blocking peptide strategies identified two domains that could account for SV binding; one in an alternatively spliced region (E44) and a second more distal site. Our findings provide a molecular basis for CaV2.1 SV binding that can account for recent evidence of C-terminal-dependent transmitter release modulation and that may contribute to SV tethering within the CaV2.1 single channel Ca2+ domain.
Collapse
Affiliation(s)
- Christine A Snidal
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Qi Li
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Brittany B Elliott
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Henry K-H Mah
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Robert H C Chen
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sabiha R Gardezi
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Elise F Stanley
- Presynaptic Mechanisms Laboratory, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
48
|
Thalhammer A, Contestabile A, Ermolyuk YS, Ng T, Volynski KE, Soong TW, Goda Y, Cingolani LA. Alternative Splicing of P/Q-Type Ca 2+ Channels Shapes Presynaptic Plasticity. Cell Rep 2018; 20:333-343. [PMID: 28700936 DOI: 10.1016/j.celrep.2017.06.055] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 06/04/2017] [Accepted: 06/21/2017] [Indexed: 12/26/2022] Open
Abstract
Alternative splicing of pre-mRNAs is prominent in the mammalian brain, where it is thought to expand proteome diversity. For example, alternative splicing of voltage-gated Ca2+ channel (VGCC) α1 subunits can generate thousands of isoforms with differential properties and expression patterns. However, the impact of this molecular diversity on brain function, particularly on synaptic transmission, which crucially depends on VGCCs, is unclear. Here, we investigate how two major splice isoforms of P/Q-type VGCCs (Cav2.1[EFa/b]) regulate presynaptic plasticity in hippocampal neurons. We find that the efficacy of P/Q-type VGCC isoforms in supporting synaptic transmission is markedly different, with Cav2.1[EFa] promoting synaptic depression and Cav2.1[EFb] synaptic facilitation. Following a reduction in network activity, hippocampal neurons upregulate selectively Cav2.1[EFa], the isoform exhibiting the higher synaptic efficacy, thus effectively supporting presynaptic homeostatic plasticity. Therefore, the balance between VGCC splice variants at the synapse is a key factor in controlling neurotransmitter release and presynaptic plasticity.
Collapse
Affiliation(s)
- Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova 16132, Italy
| | - Andrea Contestabile
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | | | - Teclise Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | | | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Yukiko Goda
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Lorenzo A Cingolani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova 16132, Italy.
| |
Collapse
|
49
|
de Jong APH, Roggero CM, Ho MR, Wong MY, Brautigam CA, Rizo J, Kaeser PS. RIM C 2B Domains Target Presynaptic Active Zone Functions to PIP 2-Containing Membranes. Neuron 2018; 98:335-349.e7. [PMID: 29606581 DOI: 10.1016/j.neuron.2018.03.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/01/2018] [Accepted: 03/05/2018] [Indexed: 11/16/2022]
Abstract
Rapid and efficient synaptic vesicle fusion requires a pool of primed vesicles, the nearby tethering of Ca2+ channels, and the presence of the phospholipid PIP2 in the target membrane. Although the presynaptic active zone mediates the first two requirements, it is unclear how fusion is targeted to membranes with high PIP2 content. Here we find that the C2B domain of the active zone scaffold RIM is critical for action potential-triggered fusion. Remarkably, the known RIM functions in vesicle priming and Ca2+ influx do not require RIM C2B domains. Instead, biophysical experiments reveal that RIM C2 domains, which lack Ca2+ binding, specifically bind to PIP2. Mutational analyses establish that PIP2 binding to RIM C2B and its tethering to the other RIM domains are crucial for efficient exocytosis. We propose that RIM C2B domains are constitutive PIP2-binding modules that couple mechanisms for vesicle priming and Ca2+ channel tethering to PIP2-containing target membranes.
Collapse
Affiliation(s)
- Arthur P H de Jong
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos M Roggero
- Departments of Biophysics, Biochemistry, and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Meng-Ru Ho
- Departments of Biophysics, Biochemistry, and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Man Yan Wong
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Chad A Brautigam
- Departments of Biophysics and Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Josep Rizo
- Departments of Biophysics, Biochemistry, and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Gandasi NR, Yin P, Omar-Hmeadi M, Ottosson Laakso E, Vikman P, Barg S. Glucose-Dependent Granule Docking Limits Insulin Secretion and Is Decreased in Human Type 2 Diabetes. Cell Metab 2018; 27:470-478.e4. [PMID: 29414688 DOI: 10.1016/j.cmet.2017.12.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 12/23/2017] [Indexed: 01/19/2023]
Abstract
Glucose-stimulated insulin secretion is biphasic, with a rapid first phase and a slowly developing sustained second phase; both are disturbed in type 2 diabetes (T2D). Biphasic secretion results from vastly different release probabilities of individual insulin granules, but the morphological and molecular basis for this is unclear. Here, we show that human insulin secretion and exocytosis critically depend on the availability of membrane-docked granules and that T2D is associated with a strong reduction in granule docking. Glucose accelerated granule docking, and this effect was absent in T2D. Newly docked granules only slowly acquired release competence; this was regulated by major signaling pathways, but not glucose. Gene expression analysis indicated that key proteins involved in granule docking are downregulated in T2D, and overexpression of these proteins increased granule docking. The findings establish granule docking as an important glucose-dependent step in human insulin secretion that is dysregulated in T2D.
Collapse
Affiliation(s)
- Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Peng Yin
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Emilia Ottosson Laakso
- Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 20502 Malmö, Sweden
| | - Petter Vikman
- Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 20502 Malmö, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden.
| |
Collapse
|