1
|
Conti R, Auger C. Associative plasticity of granule cell inputs to cerebellar Purkinje cells. eLife 2024; 13:RP96140. [PMID: 39660722 PMCID: PMC11634063 DOI: 10.7554/elife.96140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Granule cells of the cerebellum make up to 175,000 excitatory synapses on a single Purkinje cell, encoding the wide variety of information from the mossy fibre inputs into the cerebellar cortex. The granule cell axon is made of an ascending portion and a long parallel fibre extending at right angles, an architecture suggesting that synapses formed by the two segments of the axon could encode different information. There are controversial indications that ascending axon (AA) and parallel fibre (PF) synapse properties and modalities of plasticity are different. We tested the hypothesis that AA and PF synapses encode different information, and that the association of these distinct inputs to Purkinje cells might be relevant to the circuit and trigger plasticity, similar to the coincident activation of PF and climbing fibre inputs. Here, by recording synaptic currents in Purkinje cells from either proximal or distal granule cells (mostly AA and PF synapses, respectively), we describe a new form of associative plasticity between these two distinct granule cell inputs. We show for the first time that synchronous AA and PF repetitive train stimulation, with inhibition intact, triggers long-term potentiation (LTP) at AA synapses specifically. Furthermore, the timing of the presentation of the two inputs controls the outcome of plasticity and induction requires NMDAR and mGluR1 activation. The long length of the PFs allows us to preferentially activate the two inputs independently, and despite a lack of morphological reconstruction of the connections, these observations reinforce the suggestion that AA and PF synapses have different coding capabilities and plasticity that is associative, enabling effective association of information transmitted via granule cells.
Collapse
Affiliation(s)
- Rossella Conti
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the NeurosciencesParisFrance
| | - Céline Auger
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the NeurosciencesParisFrance
| |
Collapse
|
2
|
Lin TF, Busch SE, Hansel C. Intrinsic and synaptic determinants of receptive field plasticity in Purkinje cells of the mouse cerebellum. Nat Commun 2024; 15:4645. [PMID: 38821918 PMCID: PMC11143328 DOI: 10.1038/s41467-024-48373-3] [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: 08/15/2023] [Accepted: 04/28/2024] [Indexed: 06/02/2024] Open
Abstract
Non-synaptic (intrinsic) plasticity of membrane excitability contributes to aspects of memory formation, but it remains unclear whether it merely facilitates synaptic long-term potentiation or plays a permissive role in determining the impact of synaptic weight increase. We use tactile stimulation and electrical activation of parallel fibers to probe intrinsic and synaptic contributions to receptive field plasticity in awake mice during two-photon calcium imaging of cerebellar Purkinje cells. Repetitive activation of both stimuli induced response potentiation that is impaired in mice with selective deficits in either synaptic or intrinsic plasticity. Spatial analysis of calcium signals demonstrated that intrinsic, but not synaptic plasticity, enhances the spread of dendritic parallel fiber response potentiation. Simultaneous dendrite and axon initial segment recordings confirm these dendritic events affect axonal output. Our findings support the hypothesis that intrinsic plasticity provides an amplification mechanism that exerts a permissive control over the impact of long-term potentiation on neuronal responsiveness.
Collapse
Affiliation(s)
- Ting-Feng Lin
- Department of Neurobiology and Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Silas E Busch
- Department of Neurobiology and Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Christian Hansel
- Department of Neurobiology and Neuroscience Institute, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Canepari M, Ross WN. Spatial and temporal aspects of neuronal calcium and sodium signals measured with low-affinity fluorescent indicators. Pflugers Arch 2024; 476:39-48. [PMID: 37798555 DOI: 10.1007/s00424-023-02865-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023]
Abstract
Low-affinity fluorescent indicators for Ca2+ or Na+ allow measuring the dynamics of intracellular concentration of these ions with little perturbation from physiological conditions because they are weak buffers. When using synthetic indicators, which are small molecules with fast kinetics, it is also possible to extract spatial and temporal information on the sources of ion transients, their localization, and their disposition. This review examines these important aspects from the biophysical point of view, and how they have been recently exploited in neurophysiological studies. We first analyze the environment where Ca2+ and Na+ indicators are inserted, highlighting the interpretation of the two different signals. Then, we address the information that can be obtained by analyzing the rising phase and the falling phase of the Ca2+ and Na+ transients evoked by different stimuli, focusing on the kinetics of ionic currents and on the spatial interpretation of these measurements, especially on events in axons and dendritic spines. Finally, we suggest how Ca2+ or Na+ imaging using low-affinity synthetic fluorescent indicators can be exploited in future fundamental or applied research.
Collapse
Affiliation(s)
- Marco Canepari
- LIPhy, CNRS, Univ. Grenoble Alpes, F-38000, Grenoble, France.
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Valbonne, France.
- Institut National de la Santé et Recherche Médicale, Paris, France.
| | - William N Ross
- Department of Physiology, New York Medical College, Valhalla, NY, 10595, USA
| |
Collapse
|
4
|
Yang X, Liu Q, Lai MF, Ma XH, Hao XT, Xu JJ, Guo WJ. Case report: Orthostatic leg tremor as the initial manifestation in a patient with metabotropic glutamate receptor-5 encephalitis without cortical dysfunction: complexities in identification and treatment. Front Neurol 2023; 14:1288075. [PMID: 38162450 PMCID: PMC10755007 DOI: 10.3389/fneur.2023.1288075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Objective Metabotropic glutamate receptor 5 (mGluR5) encephalitis is such a rare type of autoimmune encephalitis that its diagnosis remains a challenge. Case report A 19-year-old female patient initially presented with anxiety and orthostatic leg tremors without cortical dysfunction. We found that this patient was positive for mGluR5 antibodies in both serum (1:1,000) and cerebrospinal fluid (1:32). After comprehensive intervention, the patient showed complete recovery at the 18-month follow-up. Discussion This report expands our knowledge of the possible presentations of mGluR5 encephalitis for early diagnosis, which makes it possible to prevent serious consequences and improve the prognosis.
Collapse
Affiliation(s)
- Xia Yang
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qiong Liu
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ming-feng Lai
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiao-hong Ma
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Psychiatric Laboratory and Mental Health Center, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiao-ting Hao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Jia-jun Xu
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wan-jun Guo
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou Seventh People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Lin TF, Busch SE, Hansel C. Intrinsic and synaptic determinants of receptive field plasticity in Purkinje cells of the mouse cerebellum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549760. [PMID: 37502848 PMCID: PMC10370111 DOI: 10.1101/2023.07.19.549760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Non-synaptic ('intrinsic') plasticity of membrane excitability contributes to aspects of memory formation, but it remains unclear whether it merely facilitates synaptic long-term potentiation (LTP), or whether it plays a permissive role in determining the impact of synaptic weight increase. We use tactile stimulation and electrical activation of parallel fibers to probe intrinsic and synaptic contributions to receptive field (RF) plasticity in awake mice during two-photon calcium imaging of cerebellar Purkinje cells. Repetitive activation of both stimuli induced response potentiation that is impaired in mice with selective deficits in either intrinsic plasticity (SK2 KO) or LTP (CaMKII TT305/6VA). Intrinsic, but not synaptic, plasticity expands the local, dendritic RF representation. Simultaneous dendrite and axon initial segment recordings confirm that these dendritic events affect axonal output. Our findings support the hypothesis that intrinsic plasticity provides an amplification mechanism that exerts a permissive control over the impact of LTP on neuronal responsiveness.
Collapse
Affiliation(s)
- Ting-Feng Lin
- Department of Neurobiology and Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Silas E Busch
- Department of Neurobiology and Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Christian Hansel
- Department of Neurobiology and Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
6
|
Servettini I, Talani G, Megaro A, Setzu MD, Biggio F, Briffa M, Guglielmi L, Savalli N, Binda F, Delicata F, Bru–Mercier G, Vassallo N, Maglione V, Cauchi RJ, Di Pardo A, Collu M, Imbrici P, Catacuzzeno L, D’Adamo MC, Olcese R, Pessia M. An activator of voltage-gated K + channels Kv1.1 as a therapeutic candidate for episodic ataxia type 1. Proc Natl Acad Sci U S A 2023; 120:e2207978120. [PMID: 37487086 PMCID: PMC10401004 DOI: 10.1073/pnas.2207978120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Loss-of-function mutations in the KCNA1(Kv1.1) gene cause episodic ataxia type 1 (EA1), a neurological disease characterized by cerebellar dysfunction, ataxic attacks, persistent myokymia with painful cramps in skeletal muscles, and epilepsy. Precision medicine for EA1 treatment is currently unfeasible, as no drug that can enhance the activity of Kv1.1-containing channels and offset the functional defects caused by KCNA1 mutations has been clinically approved. Here, we uncovered that niflumic acid (NFA), a currently prescribed analgesic and anti-inflammatory drug with an excellent safety profile in the clinic, potentiates the activity of Kv1.1 channels. NFA increased Kv1.1 current amplitudes by enhancing the channel open probability, causing a hyperpolarizing shift in the voltage dependence of both channel opening and gating charge movement, slowing the OFF-gating current decay. NFA exerted similar actions on both homomeric Kv1.2 and heteromeric Kv1.1/Kv1.2 channels, which are formed in most brain structures. We show that through its potentiating action, NFA mitigated the EA1 mutation-induced functional defects in Kv1.1 and restored cerebellar synaptic transmission, Purkinje cell availability, and precision of firing. In addition, NFA ameliorated the motor performance of a knock-in mouse model of EA1 and restored the neuromuscular transmission and climbing ability in Shaker (Kv1.1) mutant Drosophila melanogaster flies (Sh5). By virtue of its multiple actions, NFA has strong potential as an efficacious single-molecule-based therapeutic agent for EA1 and serves as a valuable model for drug discovery.
Collapse
Affiliation(s)
- Ilenio Servettini
- Section of Physiology, Department of Medicine, University of Perugia, Perugia06123, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council, Monserrato09042, Italy
| | - Alfredo Megaro
- Section of Physiology, Department of Medicine, University of Perugia, Perugia06123, Italy
| | - Maria Dolores Setzu
- Department of Biomedical Sciences, University of Cagliari, Monserrato09042, Italy
| | - Francesca Biggio
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato09042, Italy
| | - Michelle Briffa
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MsidaMSD2080, Malta
| | - Luca Guglielmi
- Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Nicoletta Savalli
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Francesca Binda
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne 1011, Switzerland
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, StrasbourgF-67000, France
| | - Francis Delicata
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MBR3E 0T5, Canada
| | - Gilles Bru–Mercier
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain17666, United Arab Emirates
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MsidaMSD2080, Malta
| | - Vittorio Maglione
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Pozzilli86077, Italy
| | - Ruben J. Cauchi
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MsidaMSD2080, Malta
| | - Alba Di Pardo
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Pozzilli86077, Italy
| | - Maria Collu
- Department of Biomedical Sciences, University of Cagliari, Monserrato09042, Italy
| | - Paola Imbrici
- Department of Pharmacy–Drug Sciences, University of Bari ‘‘Aldo Moro”, 70125Bari, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia06123, Italy
| | - Maria Cristina D’Adamo
- Department of Medicine and Surgery, Libera Università Mediterranea ‘‘Giuseppe DEGENNARO”, Casamassima 70010, Italy
| | - Riccardo Olcese
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MsidaMSD2080, Malta
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain17666, United Arab Emirates
| |
Collapse
|
7
|
Mansouri M, Kremser L, Nguyen TP, Kasugai Y, Caberlotto L, Gassmann M, Sarg B, Lindner H, Bettler B, Carboni L, Ferraguti F. Protein Networks Associated with Native Metabotropic Glutamate 1 Receptors (mGlu 1) in the Mouse Cerebellum. Cells 2023; 12:1325. [PMID: 37174725 PMCID: PMC10177021 DOI: 10.3390/cells12091325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
The metabotropic glutamate receptor 1 (mGlu1) plays a pivotal role in synaptic transmission and neuronal plasticity. Despite the fact that several interacting proteins involved in the mGlu1 subcellular trafficking and intracellular transduction mechanisms have been identified, the protein network associated with this receptor in specific brain areas remains largely unknown. To identify novel mGlu1-associated protein complexes in the mouse cerebellum, we used an unbiased tissue-specific proteomic approach, namely co-immunoprecipitation followed by liquid chromatography/tandem mass spectrometry analysis. Many well-known protein complexes as well as novel interactors were identified, including G-proteins, Homer, δ2 glutamate receptor, 14-3-3 proteins, and Na/K-ATPases. A novel putative interactor, KCTD12, was further investigated. Reverse co-immunoprecipitation with anti-KCTD12 antibodies revealed mGlu1 in wild-type but not in KCTD12-knock-out homogenates. Freeze-fracture replica immunogold labeling co-localization experiments showed that KCTD12 and mGlu1 are present in the same nanodomain in Purkinje cell spines, although at a distance that suggests that this interaction is mediated through interposed proteins. Consistently, mGlu1 could not be co-immunoprecipitated with KCTD12 from a recombinant mammalian cell line co-expressing the two proteins. The possibility that this interaction was mediated via GABAB receptors was excluded by showing that mGlu1 and KCTD12 still co-immunoprecipitated from GABAB receptor knock-out tissue. In conclusion, this study identifies tissue-specific mGlu1-associated protein clusters including KCTD12 at Purkinje cell synapses.
Collapse
Affiliation(s)
- Mahnaz Mansouri
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.M.); (Y.K.)
| | - Leopold Kremser
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.K.); (B.S.); (H.L.)
| | | | - Yu Kasugai
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.M.); (Y.K.)
| | - Laura Caberlotto
- Centre for Computational and Systems Biology (COSBI), The Microsoft Research University of Trento, 38068 Rovereto, Italy;
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland; (M.G.); (B.B.)
| | - Bettina Sarg
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.K.); (B.S.); (H.L.)
| | - Herbert Lindner
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.K.); (B.S.); (H.L.)
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland; (M.G.); (B.B.)
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.M.); (Y.K.)
| |
Collapse
|
8
|
Correa BH, Moreira CR, Hildebrand ME, Vieira LB. The Role of Voltage-Gated Calcium Channels in Basal Ganglia Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:183-201. [PMID: 35339179 PMCID: PMC10190140 DOI: 10.2174/1570159x20666220327211156] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 03/14/2022] [Indexed: 11/22/2022] Open
Abstract
Calcium (Ca2+) plays a central role in regulating many cellular processes and influences cell survival. Several mechanisms can disrupt Ca2+ homeostasis to trigger cell death, including oxidative stress, mitochondrial damage, excitotoxicity, neuroinflammation, autophagy, and apoptosis. Voltage-gated Ca2+ channels (VGCCs) act as the main source of Ca2+ entry into electrically excitable cells, such as neurons, and they are also expressed in glial cells such as astrocytes and oligodendrocytes. The dysregulation of VGCC activity has been reported in both Parkinson's disease (PD) and Huntington's (HD). PD and HD are progressive neurodegenerative disorders (NDs) of the basal ganglia characterized by motor impairment as well as cognitive and psychiatric dysfunctions. This review will examine the putative role of neuronal VGCCs in the pathogenesis and treatment of central movement disorders, focusing on PD and HD. The link between basal ganglia disorders and VGCC physiology will provide a framework for understanding the neurodegenerative processes that occur in PD and HD, as well as a possible path towards identifying new therapeutic targets for the treatment of these debilitating disorders.
Collapse
Affiliation(s)
- Bernardo H.M. Correa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlos Roberto Moreira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
9
|
Surdin T, Preissing B, Rohr L, Grömmke M, Böke H, Barcik M, Azimi Z, Jancke D, Herlitze S, Mark MD, Siveke I. Optogenetic activation of mGluR1 signaling in the cerebellum induces synaptic plasticity. iScience 2022; 26:105828. [PMID: 36632066 PMCID: PMC9826949 DOI: 10.1016/j.isci.2022.105828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/21/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Neuronal plasticity underlying cerebellar learning behavior is strongly associated with type 1 metabotropic glutamate receptor (mGluR1) signaling. Activation of mGluR1 leads to activation of the Gq/11 pathway, which is involved in inducing synaptic plasticity at the parallel fiber-Purkinje cell synapse (PF-PC) in form of long-term depression (LTD). To optogenetically modulate mGluR1 signaling we fused mouse melanopsin (OPN4) that activates the Gq/11 pathway to the C-termini of mGluR1 splice variants (OPN4-mGluR1a and OPN4-mGluR1b). Activation of both OPN4-mGluR1 variants showed robust Ca2+ increase in HEK cells and PCs of cerebellar slices. We provide the prove-of-concept approach to modulate synaptic plasticity via optogenetic activation of OPN4-mGluR1a inducing LTD at the PF-PC synapse in vitro. Moreover, we demonstrate that light activation of mGluR1a signaling pathway by OPN4-mGluR1a in PCs leads to an increase in intrinsic activity of PCs in vivo and improved cerebellum driven learning behavior.
Collapse
Affiliation(s)
- Tatjana Surdin
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Bianca Preissing
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Lennard Rohr
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Michelle Grömmke
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Hanna Böke
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Maike Barcik
- Cardiovascular Research Institute Düsseldorf, Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Zohre Azimi
- Optical Imaging Group, Institut für Neuroinformatik, Ruhr-University Bochum, Bochum, Germany
| | - Dirk Jancke
- Optical Imaging Group, Institut für Neuroinformatik, Ruhr-University Bochum, Bochum, Germany
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany,Corresponding author
| | - Melanie D. Mark
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Ida Siveke
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany,Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany,Corresponding author
| |
Collapse
|
10
|
Eickhoff A, Tjaden J, Stahlke S, Vorgerd M, Theis V, Matschke V, Theiss C. Effects of progesterone on T-type-Ca 2+-channel expression in Purkinje cells. Neural Regen Res 2022; 17:2465-2471. [PMID: 35535898 PMCID: PMC9120685 DOI: 10.4103/1673-5374.339008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Plasticity of cerebellar Purkinje cells (PC) is influenced by progesterone via the classical progesterone receptors PR-A and PR-B by stimulating dendritogenesis, spinogenesis, and synaptogenesis in these cells. Dissociated PC cultures were used to analyze progesterone effects at a molecular level on the voltage-gated T-type-Ca2+-channels Cav3.1, Cav3.2, and Cav3.3 as they helped determine neuronal plasticity by regulating Ca2+-influx in neuronal cells. The results showed direct effects of progesterone on the mRNA expression of T-type-Ca2+-channels, as well as on the protein kinases A and C being involved in downstream signaling pathways that play an important role in neuronal plasticity. For the mRNA expression studies of T-type-Ca2+-channels and protein kinases of the signaling cascade, laser microdissection and purified PC cultures of different maturation stages were used. Immunohistochemical staining was also performed to characterize the localization of T-type-Ca2+-channels in PC. Experimental progesterone treatment was performed on the purified PC culture for 24 and 48 hours. Our results show that progesterone increases the expression of Cav3.1 and Cav3.3 and associated protein kinases A and C in PC at the mRNA level within 48 hours after treatment at latest. These effects extend the current knowledge of the function of progesterone in the central nervous system and provide an explanatory approach for its influence on neuronal plasticity.
Collapse
Affiliation(s)
- Annika Eickhoff
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Jonas Tjaden
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil, Ruhr-Universität Bochum, Bochum, Germany
| | - Verena Theis
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
11
|
Dorgans K, Guo D, Kurima K, Wickens J, Uusisaari MY. Designing AAV Vectors for Monitoring the Subtle Calcium Fluctuations of Inferior Olive Network in vivo. Front Cell Neurosci 2022; 16:825056. [PMID: 35573836 PMCID: PMC9093741 DOI: 10.3389/fncel.2022.825056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated viral (AAV) vectors, used as vehicles for gene transfer into the brain, are a versatile and powerful tool of modern neuroscience that allow identifying specific neuronal populations, monitoring and modulating their activity. For consistent and reproducible results, the AAV vectors must be engineered so that they reliably and accurately target cell populations. Furthermore, transgene expression must be adjusted to sufficient and safe levels compatible with the physiology of studied cells. We undertook the effort to identify and validate an AAV vector that could be utilized for researching the inferior olivary (IO) nucleus, a structure gating critical timing-related signals to the cerebellum. By means of systematic construct generation and quantitative expression profiling, we succeeded in creating a viral tool for specific and strong transfection of the IO neurons without adverse effects on their physiology. The potential of these tools is demonstrated by expressing the calcium sensor GCaMP6s in adult mouse IO neurons. We could monitor subtle calcium fluctuations underlying two signatures of intrinsic IO activity: the subthreshold oscillations (STOs) and the variable-duration action potential waveforms both in-vitro and in-vivo. Further, we show that the expression levels of GCaMP6s allowing such recordings are compatible with the delicate calcium-based dynamics of IO neurons, inviting future work into the network dynamics of the olivo-cerebellar system in behaving animals.
Collapse
Affiliation(s)
- Kevin Dorgans
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Da Guo
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kiyoto Kurima
- Neurobiology Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jeff Wickens
- Neurobiology Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marylka Yoe Uusisaari
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- *Correspondence: Marylka Yoe Uusisaari
| |
Collapse
|
12
|
Hampe CS, Mitoma H. A Breakdown of Immune Tolerance in the Cerebellum. Brain Sci 2022; 12:brainsci12030328. [PMID: 35326284 PMCID: PMC8946792 DOI: 10.3390/brainsci12030328] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cerebellar dysfunction can be associated with ataxia, dysarthria, dysmetria, nystagmus and cognitive deficits. While cerebellar dysfunction can be caused by vascular, traumatic, metabolic, genetic, inflammatory, infectious, and neoplastic events, the cerebellum is also a frequent target of autoimmune attacks. The underlying cause for this vulnerability is unclear, but it may be a result of region-specific differences in blood–brain barrier permeability, the high concentration of neurons in the cerebellum and the presence of autoantigens on Purkinje cells. An autoimmune response targeting the cerebellum—or any structure in the CNS—is typically accompanied by an influx of peripheral immune cells to the brain. Under healthy conditions, the brain is protected from the periphery by the blood–brain barrier, blood–CSF barrier, and blood–leptomeningeal barrier. Entry of immune cells to the brain for immune surveillance occurs only at the blood-CSF barrier and is strictly controlled. A breakdown in the barrier permeability allows peripheral immune cells uncontrolled access to the CNS. Often—particularly in infectious diseases—the autoimmune response develops because of molecular mimicry between the trigger and a host protein. In this review, we discuss the immune surveillance of the CNS in health and disease and also discuss specific examples of autoimmunity affecting the cerebellum.
Collapse
Affiliation(s)
- Christiane S. Hampe
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Correspondence: ; Tel.: +1-206-554-9181
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo 160-0023, Japan;
| |
Collapse
|
13
|
Update on Paraneoplastic Cerebellar Degeneration. Brain Sci 2021; 11:brainsci11111414. [PMID: 34827413 PMCID: PMC8615604 DOI: 10.3390/brainsci11111414] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Purpose of review: To provide an update on paraneoplastic cerebellar degeneration (PCD), the involved antibodies and tumors, as well as management strategies. Recent findings: PCD represents the second most common presentation of the recently established class of immune mediated cerebellar ataxias (IMCAs). Although rare in general, PCD is one of the most frequent paraneoplastic presentations and characterized clinically by a rapidly progressive cerebellar syndrome. In recent years, several antibodies have been described in association with the clinical syndrome related to PCD; their clinical significance, however, has yet to be determined. The 2021 updated diagnostic criteria for paraneoplastic neurologic symptoms help to establish the diagnosis of PCD, direct cancer screening, and to evaluate the presence of these newly identified antibodies. Recognition of the clinical syndrome and prompt identification of a specific antibody are essential for early detection of an underlying malignancy and initiation of an appropriate treatment, which represents the best opportunity to modulate the course of the disease. As clinical symptoms can precede tumor diagnosis by years, co-occurrence of specific symptoms and antibodies should prompt continuous surveillance of the patient. Summary: We provide an in-depth overview on PCD, summarize recent findings related to PCD, and highlight the transformed diagnostic approach.
Collapse
|
14
|
Shao J, Liu Y, Gao D, Tu J, Yang F. Neural Burst Firing and Its Roles in Mental and Neurological Disorders. Front Cell Neurosci 2021; 15:741292. [PMID: 34646123 PMCID: PMC8502892 DOI: 10.3389/fncel.2021.741292] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022] Open
Abstract
Neural firing patterns are critical for specific information coding and transmission, and abnormal firing is implicated in a series of neural pathologies. Recent studies have indicated that enhanced burst firing mediated by T-type voltage-gated calcium channels (T-VGCCs) in specific neuronal subtypes is involved in several mental or neurological disorders such as depression and epilepsy, while suppression of T-VGCCs relieve related symptoms. Burst firing consists of groups of relatively high-frequency spikes separated by quiescence. Neurons in a variety of brain areas, including the thalamus, hypothalamus, cortex, and hippocampus, display burst firing, but the ionic mechanisms that generating burst firing and the related physiological functions vary among regions. In this review, we summarize recent findings on the mechanisms underlying burst firing in various brain areas, as well as the roles of burst firing in several mental and neurological disorders. We also discuss the ion channels and receptors that may regulate burst firing directly or indirectly, with these molecules highlighted as potential intervention targets for the treatment of mental and neurological disorders.
Collapse
Affiliation(s)
- Jie Shao
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Yunhui Liu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Dashuang Gao
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Tu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Fan Yang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Tjaden J, Eickhoff A, Stahlke S, Gehmeyr J, Vorgerd M, Theis V, Matschke V, Theiss C. Expression Pattern of T-Type Ca 2+ Channels in Cerebellar Purkinje Cells after VEGF Treatment. Cells 2021; 10:2277. [PMID: 34571926 PMCID: PMC8470219 DOI: 10.3390/cells10092277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 12/04/2022] Open
Abstract
T-type Ca2+ channels, generating low threshold calcium influx in neurons, play a crucial role in the function of neuronal networks and their plasticity. To further investigate their role in the complex field of research in plasticity of neurons on a molecular level, this study aimed to analyse the impact of the vascular endothelial growth factor (VEGF) on these channels. VEGF, known as a player in vasculogenesis, also shows potent influence in the central nervous system, where it elicits neuronal growth. To investigate the influence of VEGF on the three T-type Ca2+ channel isoforms, Cav3.1 (encoded by Cacna1g), Cav3.2 (encoded by Cacna1h), and Cav3.3 (encoded by Cacna1i), lasermicrodissection of in vivo-grown Purkinje cells (PCs) was performed, gene expression was analysed via qPCR and compared to in vitro-grown PCs. We investigated the VEGF receptor composition of in vivo- and in vitro-grown PCs and underlined the importance of VEGF receptor 2 for PCs. Furthermore, we performed immunostaining of T-type Ca2+ channels with in vivo- and in vitro-grown PCs and showed the distribution of T-type Ca2+ channel expression during PC development. Overall, our findings provide the first evidence that the mRNA expression of Cav3.1, Cav3.2, and Cav3.3 increases due to VEGF stimulation, which indicates an impact of VEGF on neuronal plasticity.
Collapse
Affiliation(s)
- Jonas Tjaden
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| | - Annika Eickhoff
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| | - Julian Gehmeyr
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| | - Matthias Vorgerd
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil, Ruhr-University Bochum, Buerkle-de-la-Camp-Platz 1, 44789 Bochum, Germany;
| | - Verena Theis
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Universitätsstr. 150, 44801 Bochum, Germany; (J.T.); (A.E.); (S.S.); (J.G.); (V.T.); (V.M.)
| |
Collapse
|
16
|
mGluR1 signaling in cerebellar Purkinje cells: Subcellular organization and involvement in cerebellar function and disease. Neuropharmacology 2021; 194:108629. [PMID: 34089728 DOI: 10.1016/j.neuropharm.2021.108629] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/20/2022]
Abstract
The cerebellum is essential for the control, coordination, and learning of movements, and for certain aspects of cognitive function. Purkinje cells are the sole output neurons in the cerebellar cortex and therefore play crucial roles in the diverse functions of the cerebellum. The type 1 metabotropic glutamate receptor (mGluR1) is prominently enriched in Purkinje cells and triggers downstream signaling pathways that are required for functional and structural plasticity, and for synaptic responses. To understand how mGluR1 contributes to cerebellar functions, it is important to consider not only the operational properties of this receptor, but also its spatial organization and the molecular interactions that enable its proper functioning. In this review, we highlight how mGluR1 and its related signaling molecules are organized into tightly coupled microdomains to fulfill physiological functions. We also describe emerging evidence that altered mGluR1 signaling in Purkinje cells underlies cerebellar dysfunction in ataxias of human patients and mouse models.
Collapse
|
17
|
Harding EK, Dedek A, Bonin RP, Salter MW, Snutch TP, Hildebrand ME. The T-type calcium channel antagonist, Z944, reduces spinal excitability and pain hypersensitivity. Br J Pharmacol 2021; 178:3517-3532. [PMID: 33871884 PMCID: PMC8453510 DOI: 10.1111/bph.15498] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/02/2021] [Accepted: 04/05/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose T‐type voltage‐gated calcium channels are an emerging therapeutic target for neurological disorders including epilepsy and pain. Inhibition of T‐type channels reduces the excitability of peripheral nociceptive sensory neurons and reverses pain hypersensitivity in male rodent pain models. However, administration of peripherally restricted T‐type antagonists failed to show efficacy in multiple clinical and preclinical pain trials, suggesting that inhibition of peripheral T‐type channels alone may be insufficient for pain relief. Experimental Approach We utilized the selective and CNS‐penetrant T‐type channel antagonist, Z944, in electrophysiological, calcium imaging and behavioural paradigms to determine its effect on lamina I neuron excitability and inflammatory pain behaviours. Key Results Voltage‐clamp recordings from lamina I spinal neurons of adult rats revealed that approximately 80% of neurons possess a low threshold T‐type current, which was blocked by Z944. Due to this highly prevalent T‐type current, Z944 potently blocked action‐potential evoked somatic and dendritic calcium transients in lamina I neurons. Moreover, application of Z944 to spinal cord slices attenuated action potential firing rates in over half of laminae I/II neurons. Finally, we found that intraperitoneal injection of Z944 (1–10 mg·kg−1) dose‐dependently reversed mechanical allodynia in the complete Freund's adjuvant model of persistent inflammatory pain, with a similar magnitude and time course of analgesic effects between male and female rats. Conclusion and Implications T‐type calcium channels critically shape the excitability of lamina I pain processing neurons and inhibition of these channels by the clinical stage antagonist Z944 potently reverses pain hypersensitivity across sexes.
Collapse
Affiliation(s)
- Erika K Harding
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annemarie Dedek
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Robert P Bonin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,University of Toronto Centre for the Study of Pain, University of Toronto, Toronto, Ontario, Canada
| | - Michael W Salter
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael E Hildebrand
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Roome CJ, Kuhn B. Dendritic coincidence detection in Purkinje neurons of awake mice. eLife 2020; 9:59619. [PMID: 33345779 PMCID: PMC7771959 DOI: 10.7554/elife.59619] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Dendritic coincidence detection is fundamental to neuronal processing yet remains largely unexplored in awake animals. Specifically, the underlying dendritic voltage–calcium relationship has not been directly addressed. Here, using simultaneous voltage and calcium two-photon imaging of Purkinje neuron spiny dendrites, we show how coincident synaptic inputs and resulting dendritic spikes modulate dendritic calcium signaling during sensory stimulation in awake mice. Sensory stimulation increased the rate of postsynaptic potentials and dendritic calcium spikes evoked by climbing fiber and parallel fiber synaptic input. These inputs are integrated in a time-dependent and nonlinear fashion to enhance the sensory-evoked dendritic calcium signal. Intrinsic supralinear dendritic mechanisms, including voltage-gated calcium channels and metabotropic glutamate receptors, are recruited cooperatively to expand the dynamic range of sensory-evoked dendritic calcium signals. This establishes how dendrites can use multiple interplaying mechanisms to perform coincidence detection, as a fundamental and ongoing feature of dendritic integration in behaving animals.
Collapse
Affiliation(s)
- Christopher J Roome
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Bernd Kuhn
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| |
Collapse
|
19
|
Novel Missense CACNA1G Mutations Associated with Infantile-Onset Developmental and Epileptic Encephalopathy. Int J Mol Sci 2020; 21:ijms21176333. [PMID: 32878331 PMCID: PMC7503748 DOI: 10.3390/ijms21176333] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/29/2020] [Accepted: 08/29/2020] [Indexed: 11/17/2022] Open
Abstract
The CACNA1G gene encodes the low-voltage-activated Cav3.1 channel, which is expressed in various areas of the CNS, including the cerebellum. We studied two missense CACNA1G variants, p.L208P and p.L909F, and evaluated the relationships between the severity of Cav3.1 dysfunction and the clinical phenotype. The presentation was of a developmental and epileptic encephalopathy without evident cerebellar atrophy. Both patients exhibited axial hypotonia, developmental delay, and severe to profound cognitive impairment. The patient with the L909F mutation had initially refractory seizures and cerebellar ataxia, whereas the L208P patient had seizures only transiently but was overall more severely affected. In transfected mammalian cells, we determined the biophysical characteristics of L208P and L909F variants, relative to the wild-type channel and a previously reported gain-of-function Cav3.1 variant. The L208P mutation shifted the activation and inactivation curves to the hyperpolarized direction, slowed the kinetics of inactivation and deactivation, and reduced the availability of Ca2+ current during repetitive stimuli. The L909F mutation impacted channel function less severely, resulting in a hyperpolarizing shift of the activation curve and slower deactivation. These data suggest that L909F results in gain-of-function, whereas L208P exhibits mixed gain-of-function and loss-of-function effects due to opposing changes in the biophysical properties. Our study expands the clinical spectrum associated with CACNA1G mutations, corroborating further the causal association with distinct complex phenotypes.
Collapse
|
20
|
Canepari M. Is Purkinje Neuron Hyperpolarisation Important for Cerebellar Synaptic Plasticity? A Retrospective and Prospective Analysis. THE CEREBELLUM 2020; 19:869-878. [PMID: 32654026 DOI: 10.1007/s12311-020-01164-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two recent studies have demonstrated that the dendritic Ca2+ signal associated with a climbing fibre (CF) input to the cerebellar Purkinje neuron (PN) depends on the membrane potential (Vm). Specifically, when the cell is hyperpolarised, this signal is mediated by T-type voltage-gated Ca2+ channels; in contrast, when the cell is firing, the CF-PN signal is mediated by P/Q-type voltage-gated Ca2+ channels. When the CF input is paired with parallel fibre (PF) activity, the signal is locally amplified at the sites of PF-activated synapses according to the Vm at the time of the CF input, suggesting that the standing Vm is a critical parameter for the induction of PF synaptic plasticity. In this review, I analyse how the Vm can potentially play a role in cerebellar learning focussing, in particular, on the hyperpolarised state that appears to occur episodically, since PNs are mostly firing under physiological conditions. By revisiting the recent literature reporting in vivo recordings and synaptic plasticity studies, I speculate on how a putative role of the PN Vm can provide an interpretation for the results of these studies.
Collapse
Affiliation(s)
- Marco Canepari
- University of Grenoble Alpes, CNRS, LIPhy, F-38000, Grenoble, France. .,Laboratories of Excellence, Ion Channel Science and Therapeutics, Valbonne, France. .,Institut National de la Santé et Recherche Médicale, Paris, France.
| |
Collapse
|
21
|
The Origin of Physiological Local mGluR1 Supralinear Ca 2+ Signals in Cerebellar Purkinje Neurons. J Neurosci 2020; 40:1795-1809. [PMID: 31969470 DOI: 10.1523/jneurosci.2406-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 11/21/2022] Open
Abstract
In mouse cerebellar Purkinje neurons (PNs), the climbing fiber (CF) input provides a signal to parallel fiber (PF) synapses, triggering PF synaptic plasticity. This signal is given by supralinear Ca2+ transients, associated with the CF synaptic potential and colocalized with the PF Ca2+ influx, occurring only when PF activity precedes the CF input. Here, we unravel the biophysical determinants of supralinear Ca2+ signals associated with paired PF-CF synaptic activity. We used membrane potential (V m) and Ca2+ imaging to investigate the local CF-associated Ca2+ influx following a train of PF synaptic potentials in two cases: (1) when the dendritic V m is hyperpolarized below the resting V m, and (2) when the dendritic V m is at rest. We found that supralinear Ca2+ signals are mediated by type-1 metabotropic glutamate receptors (mGluR1s) when the CF input is delayed by 100-150 ms from the first PF input in both cases. When the dendrite is hyperpolarized only, however, mGluR1s boost neighboring T-type channels, providing a mechanism for local coincident detection of PF-CF activity. The resulting Ca2+ elevation is locally amplified by saturation of endogenous Ca2+ buffers produced by the PF-associated Ca2+ influx via the mGluR1-mediated nonselective cation conductance. In contrast, when the dendritic V m is at rest, mGluR1s increase dendritic excitability by inactivating A-type K+ channels, but this phenomenon is not restricted to the activated PF synapses. Thus, V m is likely a crucial parameter in determining PF synaptic plasticity, and the occurrence of hyperpolarization episodes is expected to play an important role in motor learning.SIGNIFICANCE STATEMENT In Purkinje neurons, parallel fiber synaptic plasticity, determined by coincident activation of the climbing fiber input, underlies cerebellar learning. We unravel the biophysical mechanisms allowing the CF input to produce a local Ca2+ signal exclusively at the sites of activated parallel fibers. We show that when the membrane potential is hyperpolarized with respect to the resting membrane potential, type-1 metabotropic glutamate receptors locally enhance Ca2+ influx mediated by T-type Ca2+ channels, and that this signal is amplified by saturation of endogenous buffer also mediated by the same receptors. The combination of these two mechanisms is therefore capable of producing a Ca2+ signal at the activated parallel fiber sites, suggesting a role of Purkinje neuron membrane potential in cerebellar learning.
Collapse
|
22
|
Prestori F, Moccia F, D’Angelo E. Disrupted Calcium Signaling in Animal Models of Human Spinocerebellar Ataxia (SCA). Int J Mol Sci 2019; 21:ijms21010216. [PMID: 31892274 PMCID: PMC6981692 DOI: 10.3390/ijms21010216] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/22/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Spinocerebellar ataxias (SCAs) constitute a heterogeneous group of more than 40 autosomal-dominant genetic and neurodegenerative diseases characterized by loss of balance and motor coordination due to dysfunction of the cerebellum and its efferent connections. Despite a well-described clinical and pathological phenotype, the molecular and cellular events that underlie neurodegeneration are still poorly undaerstood. Emerging research suggests that mutations in SCA genes cause disruptions in multiple cellular pathways but the characteristic SCA pathogenesis does not begin until calcium signaling pathways are disrupted in cerebellar Purkinje cells. Ca2+ signaling in Purkinje cells is important for normal cellular function as these neurons express a variety of Ca2+ channels, Ca2+-dependent kinases and phosphatases, and Ca2+-binding proteins to tightly maintain Ca2+ homeostasis and regulate physiological Ca2+-dependent processes. Abnormal Ca2+ levels can activate toxic cascades leading to characteristic death of Purkinje cells, cerebellar atrophy, and ataxia that occur in many SCAs. The output of the cerebellar cortex is conveyed to the deep cerebellar nuclei (DCN) by Purkinje cells via inhibitory signals; thus, Purkinje cell dysfunction or degeneration would partially or completely impair the cerebellar output in SCAs. In the absence of the inhibitory signal emanating from Purkinje cells, DCN will become more excitable, thereby affecting the motor areas receiving DCN input and resulting in uncoordinated movements. An outstanding advantage in studying the pathogenesis of SCAs is represented by the availability of a large number of animal models which mimic the phenotype observed in humans. By mainly focusing on mouse models displaying mutations or deletions in genes which encode for Ca2+ signaling-related proteins, in this review we will discuss the several pathogenic mechanisms related to deranged Ca2+ homeostasis that leads to significant Purkinje cell degeneration and dysfunction.
Collapse
Affiliation(s)
- Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- Correspondence:
| | - Francesco Moccia
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| |
Collapse
|
23
|
Janakiraman U, Yu J, Moutal A, Chinnasamy D, Boinon L, Batchelor SN, Anandhan A, Khanna R, Nelson MA. TAF1-gene editing alters the morphology and function of the cerebellum and cerebral cortex. Neurobiol Dis 2019; 132:104539. [PMID: 31344492 PMCID: PMC7197880 DOI: 10.1016/j.nbd.2019.104539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/20/2019] [Accepted: 07/19/2019] [Indexed: 10/26/2022] Open
Abstract
TAF1/MRSX33 intellectual disability syndrome is an X-linked disorder caused by loss-of-function mutations in the TAF1 gene. How these mutations cause dysmorphology, hypotonia, intellectual and motor defects is unknown. Mouse models which have embryonically targeted TAF1 have failed, possibly due to TAF1 being essential for viability, preferentially expressed in early brain development, and intolerant of mutation. Novel animal models are valuable tools for understanding neuronal pathology. Here, we report the development and characterization of a novel animal model for TAF1 ID syndrome in which the TAF1 gene is deleted in embryonic rats using clustered regularly interspaced short palindromic repeats (CRISPR) associated protein 9 (Cas9) technology and somatic brain transgenesis mediated by lentiviral transduction. Rat pups, post-natal day 3, were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 vectors. Rats were subjected to a battery of behavioral tests followed by histopathological analyses of brains at post-natal day 14 and day 35. TAF1-edited rats exhibited behavioral deficits at both the neonatal and juvenile stages of development. Deletion of TAF1 lead to a hypoplasia and loss of the Purkinje cells. We also observed a decreased in GFAP positive astrocytes and an increase in Iba1 positive microglia within the granular layer of the cerebellum in TAF1-edited animals. Immunostaining revealed a reduction in the expression of the CaV3.1 T-type calcium channel. Abnormal motor symptoms in TAF1-edited rats were associated with irregular cerebellar output caused by changes in the intrinsic activity of the Purkinje cells due to loss of pre-synaptic CaV3.1. This animal model provides a powerful new tool for studies of neuronal dysfunction in conditions associated with TAF1 abnormalities and should prove useful for developing therapeutic strategies to treat TAF1 ID syndrome.
Collapse
Affiliation(s)
- Udaiyappan Janakiraman
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Jie Yu
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310058, China
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Dhanalakshmi Chinnasamy
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Lisa Boinon
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Shelby N Batchelor
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Annaduri Anandhan
- Department of Pharmacology and Toxicology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, United States of America; The BIO5 Institute, University of Arizona, United States of America
| | - Mark A Nelson
- Department of Pathology, University of Arizona College of Medicine and College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
24
|
Tracy ME, Tesic V, Stamenic TT, Joksimovic SM, Busquet N, Jevtovic-Todorovic V, Todorovic SM. Ca V3.1 isoform of T-type calcium channels supports excitability of rat and mouse ventral tegmental area neurons. Neuropharmacology 2018; 135:343-354. [PMID: 29578032 DOI: 10.1016/j.neuropharm.2018.03.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 10/17/2022]
Abstract
Recent data have implicated voltage-gated calcium channels in the regulation of the excitability of neurons within the mesolimbic reward system. While the attention of most research has centered on high voltage L-type calcium channel activity, the presence and role of the low voltage-gated T-type calcium channel (T-channels) has not been well explored. Hence, we investigated T-channel properties in the neurons of the ventral tegmental area (VTA) utilizing wild-type (WT) rats and mice, CaV3.1 knock-out (KO) mice, and TH-eGFP knock-in (KI) rats in acute horizontal brain slices of adolescent animals. In voltage-clamp experiments, we first assessed T-channel activity in WT rats with characteristic properties of voltage-dependent activation and inactivation, as well as characteristic crisscrossing patterns of macroscopic current kinetics. T-current kinetics were similar in WT mice and WT rats but T-currents were abolished in CaV3.1 KO mice. In ensuing current-clamp experiments, we observed the presence of hyperpolarization-induced rebound burst firing in a subset of neurons in WT rats, as well as dopaminergic and non-dopaminergic neurons in TH-eGFP KI rats. Following the application of a pan-selective T-channel blocker TTA-P2, rebound bursting was significantly inhibited in all tested cells. In a behavioral assessment, the acute locomotor increase induced by a MK-801 (Dizocilpine) injection in WT mice was abolished in CaV3.1 KO mice, suggesting a tangible role for 3.1 T-type channels in drug response. We conclude that pharmacological targeting of CaV3.1 isoform of T-channels may be a novel approach for the treatment of disorders of mesolimbic reward system.
Collapse
Affiliation(s)
- Matthew E Tracy
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, United States
| | - Vesna Tesic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, United States
| | - Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, United States
| | - Srdjan M Joksimovic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, United States
| | - Nicolas Busquet
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, United States
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, United States
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, United States; Neuroscience Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, United States.
| |
Collapse
|
25
|
Huang M, Verbeek DS. Why do so many genetic insults lead to Purkinje Cell degeneration and spinocerebellar ataxia? Neurosci Lett 2018; 688:49-57. [PMID: 29421540 DOI: 10.1016/j.neulet.2018.02.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/02/2018] [Indexed: 12/29/2022]
Abstract
The genetically heterozygous spinocerebellar ataxias are all characterized by cerebellar atrophy and pervasive Purkinje Cell degeneration. Up to date, more than 35 functionally diverse spinocerebellar ataxia genes have been identified. The main question that remains yet unsolved is why do some many genetic insults lead to Purkinje Cell degeneration and spinocerebellar ataxia? To address this question it is important to identify intrinsic pathways important for Purkinje Cell function and survival. In this review, we discuss the current consensus on shared mechanisms underlying the pervasive Purkinje Cell loss in spinocerebellar ataxia. Additionally, using recently published cell type specific expression data, we identified several Purkinje Cell-specific genes and discuss how the corresponding pathways might underlie the vulnerability of Purkinje Cells in response to the diverse genetic insults causing spinocerebellar ataxia.
Collapse
Affiliation(s)
- Miaozhen Huang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Dineke S Verbeek
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands.
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Autoantibodies to Central nervous system (CNS) metabotropic receptors are associated with a growing family of autoimmune brain diseases, including encephalitis, basal ganglia encephalitis, Ophelia syndrome, and cerebellitis. The purpose of this review is to summarize the state of knowledge regarding the target receptors, the neurological autoimmune disorders, and the pathogenic mechanisms. RECENT FINDINGS Antibodies to the γ-aminobutyric acid B receptor are associate with limbic encephalitis and severe seizures, often with small cell lung cancers. Metabotropic glutamate receptor 5 (mGluR5) antibodies associate with Ophelia syndrome, a relatively mild form of encephalitis linked to Hodgkin lymphoma. mGluR1 antibodies associate with a form of cerebellar degeneration, and also Hodgkin lymphoma. Antibodies to Homer 3, a protein associated with mGluR1, have also been reported in two patients with cerebellar syndromes. Dopamine-2 receptor antibodies have been reported by one group in children with basal ganglia encephalitis and other disorders. SUMMARY CNS metabotropic receptor antibodies may exert direct inhibitory effects on their target receptors, but the evidence is more limited than with autoantibodies to ionotropic glutamate receptors. In the future, improved recognition of these patients may lead to better outcomes. Understanding the molecular mechanisms of the diseases may uncover novel treatment strategies.
Collapse
|
27
|
Turner RW. Sweet T-bursting is the subiculum's true badge. J Physiol 2017; 595:6223. [PMID: 28786112 DOI: 10.1113/jp274981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ray W Turner
- University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
28
|
Gutierrez-Castellanos N, Da Silva-Matos CM, Zhou K, Canto CB, Renner MC, Koene LMC, Ozyildirim O, Sprengel R, Kessels HW, De Zeeuw CI. Motor Learning Requires Purkinje Cell Synaptic Potentiation through Activation of AMPA-Receptor Subunit GluA3. Neuron 2017; 93:409-424. [PMID: 28103481 PMCID: PMC5263704 DOI: 10.1016/j.neuron.2016.11.046] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 09/28/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
Abstract
Accumulating evidence indicates that cerebellar long-term potentiation (LTP) is necessary for procedural learning. However, little is known about its underlying molecular mechanisms. Whereas AMPA receptor (AMPAR) subunit rules for synaptic plasticity have been extensively studied in relation to declarative learning, it is unclear whether these rules apply to cerebellum-dependent motor learning. Here we show that LTP at the parallel-fiber-to-Purkinje-cell synapse and adaptation of the vestibulo-ocular reflex depend not on GluA1- but on GluA3-containing AMPARs. In contrast to the classic form of LTP implicated in declarative memory formation, this form of LTP does not require GluA1-AMPAR trafficking but rather requires changes in open-channel probability of GluA3-AMPARs mediated by cAMP signaling and activation of the protein directly activated by cAMP (Epac). We conclude that vestibulo-cerebellar motor learning is the first form of memory acquisition shown to depend on GluA3-dependent synaptic potentiation by increasing single-channel conductance. Cerebellar learning depends on expression of GluA3, but not GluA1, in Purkinje cells GluA3 is required to induce LTP, but not LTD, at PF-PC synapses GluA3-dependent potentiation involves a cAMP-driven change in channel conductance GluA3-mediated LTP and learning are induced via cAMP-mediated Epac activation
Collapse
Affiliation(s)
- Nicolas Gutierrez-Castellanos
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Department of Neuroscience, Erasmus MC Rotterdam, 3015 GE Rotterdam, the Netherlands
| | - Carla M Da Silva-Matos
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Kuikui Zhou
- Department of Neuroscience, Erasmus MC Rotterdam, 3015 GE Rotterdam, the Netherlands
| | - Cathrin B Canto
- Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Maria C Renner
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Linda M C Koene
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Ozgecan Ozyildirim
- Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Rolf Sprengel
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Helmut W Kessels
- Synaptic Plasticity and Behavior Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands.
| | - Chris I De Zeeuw
- Cerebellar Coordination and Cognition Group, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands; Department of Neuroscience, Erasmus MC Rotterdam, 3015 GE Rotterdam, the Netherlands
| |
Collapse
|
29
|
Kano M, Watanabe T. Type-1 metabotropic glutamate receptor signaling in cerebellar Purkinje cells in health and disease. F1000Res 2017; 6:416. [PMID: 28435670 PMCID: PMC5381626 DOI: 10.12688/f1000research.10485.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2017] [Indexed: 01/28/2023] Open
Abstract
The cerebellum is a brain structure involved in coordination, control, and learning of movements, as well as certain aspects of cognitive function. Purkinje cells are the sole output neurons from the cerebellar cortex and therefore play crucial roles in the overall function of the cerebellum. The type-1 metabotropic glutamate receptor (mGluR1) is a key “hub” molecule that is critically involved in the regulation of synaptic wiring, excitability, synaptic response, and synaptic plasticity of Purkinje cells. In this review, we aim to highlight how mGluR1 controls these events in Purkinje cells. We also describe emerging evidence that altered mGluR1 signaling in Purkinje cells underlies cerebellar dysfunctions in several clinically relevant mouse models of human ataxias.
Collapse
Affiliation(s)
- Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takaki Watanabe
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
30
|
Dalmau J, Geis C, Graus F. Autoantibodies to Synaptic Receptors and Neuronal Cell Surface Proteins in Autoimmune Diseases of the Central Nervous System. Physiol Rev 2017; 97:839-887. [PMID: 28298428 PMCID: PMC5539405 DOI: 10.1152/physrev.00010.2016] [Citation(s) in RCA: 393] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Investigations in the last 10 years have revealed a new category of neurological diseases mediated by antibodies against cell surface and synaptic proteins. There are currently 16 such diseases all characterized by autoantibodies against neuronal proteins involved in synaptic signaling and plasticity. In clinical practice these findings have changed the diagnostic and treatment approach to potentially lethal, but now treatable, neurological and psychiatric syndromes previously considered idiopathic or not even suspected to be immune-mediated. Studies show that patients' antibodies can impair the surface dynamics of the target receptors eliminating them from synapses (e.g., NMDA receptor), block the function of the antigens without changing their synaptic density (e.g., GABAb receptor), interfere with synaptic protein-protein interactions (LGI1, Caspr2), alter synapse formation (e.g., neurexin-3α), or by unclear mechanisms associate to a new form of tauopathy (IgLON5). Here we first trace the process of discovery of these diseases, describing the triggers and symptoms related to each autoantigen, and then review in detail the structural and functional alterations caused by the autoantibodies with special emphasis in those (NMDA receptor, amphiphysin) that have been modeled in animals.
Collapse
Affiliation(s)
- Josep Dalmau
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania; Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain; Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany; Servei de Neurologia, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Christian Geis
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania; Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain; Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany; Servei de Neurologia, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Graus
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania; Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain; Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany; Servei de Neurologia, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Nicholson E, Kullmann DM. T-type calcium channels contribute to NMDA receptor independent synaptic plasticity in hippocampal regular-spiking oriens-alveus interneurons. J Physiol 2017; 595:3449-3458. [PMID: 28134447 PMCID: PMC5451714 DOI: 10.1113/jp273695] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/06/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Regular-spiking interneurons in the hippocampal stratum oriens exhibit a form of long-term potentiation of excitatory transmission that is independent of NMDA receptors but requires co-activation of Ca2+ -permeable AMPA receptors and group I metabotropic glutamate receptors. We show that T-type Ca2+ channels are present in such interneurons. Blockade of T-type currents prevents the induction of long-term potentiation, and also interferes with long-lasting potentiation induced either by postsynaptic trains of action potentials or by pairing postsynaptic hyperpolarization with activation of group I metabotropic receptors. Several Ca2+ sources thus converge on the induction of NMDA receptor independent synaptic plasticity. ABSTRACT NMDA receptor independent long-term potentiation (LTP) in hippocampal stratum oriens-alveus (O/A) interneurons requires co-activation of postsynaptic group I metabotropic glutamate receptors (mGluRs) and Ca2+ -permeable AMPA receptors. The rectification properties of such AMPA receptors contribute to the preferential induction of LTP at hyperpolarized potentials. A persistent increase in excitatory transmission can also be triggered by exogenous activation of group I mGluRs at the same time as the interneuron is hyperpolarized, or by postsynaptic trains of action potentials in the absence of presynaptic stimulation. In the present study, we identify low-threshold transient (T-type) channels as a further source of Ca2+ that contributes to synaptic plasticity. T-type Ca2+ currents were detected in mouse regular-spiking O/A interneurons. Blocking T-type currents pharmacologically prevented LTP induced by high-frequency stimulation of glutamatergic axons, or by application of the group I mGluR agonist dihydroxyphenylglycine, paired with postsynaptic hyperpolarization. T-type current blockade also prevented synaptic potentiation induced by postsynaptic action potential trains. Several sources of Ca2+ thus converge on NMDA receptor independent LTP induction in O/A interneurons.
Collapse
|
32
|
Yabuki Y, Matsuo K, Izumi H, Haga H, Yoshida T, Wakamori M, Kakei A, Sakimura K, Fukuda T, Fukunaga K. Pharmacological properties of SAK3, a novel T-type voltage-gated Ca 2+ channel enhancer. Neuropharmacology 2017; 117:1-13. [PMID: 28093211 DOI: 10.1016/j.neuropharm.2017.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/27/2016] [Accepted: 01/12/2017] [Indexed: 11/30/2022]
Abstract
T-type voltage-gated Ca2+ channels (T-VGCCs) function in the pathophysiology of epilepsy, pain and sleep. However, their role in cognitive function remains unclear. We previously reported that the cognitive enhancer ST101, which stimulates T-VGCCs in rat cortical slices, was a potential Alzheimer's disease therapeutic. Here, we introduce a more potent T-VGCC enhancer, SAK3 (ethyl 8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo [1,2-a]pyridin]-2-ene-3-carboxylate), and characterize its pharmacological properties in brain. Based on whole cell patch-clamp analysis, SAK3 (0.01-10 nM) significantly enhanced Cav3.1 currents in neuro2A cells ectopically expressing Cav3.1. SAK3 (0.1-10 nM nM) also enhanced Cav3.3 but not Cav3.2 currents in the transfected cells. Notably, Cav3.1 and Cav3.3 T-VGCCs were localized in cholinergic neurve systems in hippocampus and in the medial septum. Indeed, acute oral administration of SAK3 (0.5 mg/kg, p.o.), but not ST101 (0.5 mg/kg, p.o.) significantly enhanced acetylcholine (ACh) release in the hippocampal CA1 region of naïve mice. Moreover, acute SAK3 (0.5 mg/kg, p.o.) administration significantly enhanced hippocampal ACh levels in olfactory-bulbectomized (OBX) mice, rescuing impaired memory-related behaviors. Treatment of OBX mice with the T-VGCC-specific blocker NNC 55-0396 (12.5 mg/kg, i.p.) antagonized both enhanced ACh release and memory improvements elicited by SAK3 administration. We also observed that SAK3-induced ACh releases were significantly blocked in the hippocampus from Cav3.1 knockout (KO) mice. These findings suggest overall that T-VGCCs play a key role in cognition by enhancing hippocampal ACh release and that the cognitive enhancer SAK3 could be a candidate therapeutic in Alzheimer's disease.
Collapse
Affiliation(s)
- Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hidaka Haga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takashi Yoshida
- Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Minoru Wakamori
- Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Akikazu Kakei
- Department of Chemistry and Material Engineering, Faculty of Engineering, Shinshu University, Nagano, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takaichi Fukuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
33
|
Schultz SR, Copeland CS, Foust AJ, Quicke P, Schuck R. Advances in two photon scanning and scanless microscopy technologies for functional neural circuit imaging. PROCEEDINGS OF THE IEEE. INSTITUTE OF ELECTRICAL AND ELECTRONICS ENGINEERS 2017; 105:139-157. [PMID: 28757657 PMCID: PMC5526632 DOI: 10.1109/jproc.2016.2577380] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Recent years have seen substantial developments in technology for imaging neural circuits, raising the prospect of large scale imaging studies of neural populations involved in information processing, with the potential to lead to step changes in our understanding of brain function and dysfunction. In this article we will review some key recent advances: improved fluorophores for single cell resolution functional neuroimaging using a two photon microscope; improved approaches to the problem of scanning active circuits; and the prospect of scanless microscopes which overcome some of the bandwidth limitations of current imaging techniques. These advances in technology for experimental neuroscience have in themselves led to technical challenges, such as the need for the development of novel signal processing and data analysis tools in order to make the most of the new experimental tools. We review recent work in some active topics, such as region of interest segmentation algorithms capable of demixing overlapping signals, and new highly accurate algorithms for calcium transient detection. These advances motivate the development of new data analysis tools capable of dealing with spatial or spatiotemporal patterns of neural activity, that scale well with pattern size.
Collapse
Affiliation(s)
- Simon R Schultz
- Center for Neurotechnology and Department of Bioengineering Imperial College London, South Kensington, LondonSW7 2AZ, UK
| | - Caroline S Copeland
- Center for Neurotechnology and Department of Bioengineering Imperial College London, South Kensington, LondonSW7 2AZ, UK
| | - Amanda J Foust
- Center for Neurotechnology and Department of Bioengineering Imperial College London, South Kensington, LondonSW7 2AZ, UK
| | - Peter Quicke
- Center for Neurotechnology and Department of Bioengineering Imperial College London, South Kensington, LondonSW7 2AZ, UK
| | - Renaud Schuck
- Center for Neurotechnology and Department of Bioengineering Imperial College London, South Kensington, LondonSW7 2AZ, UK
| |
Collapse
|
34
|
Ly R, Bouvier G, Szapiro G, Prosser HM, Randall AD, Kano M, Sakimura K, Isope P, Barbour B, Feltz A. Contribution of postsynaptic T-type calcium channels to parallel fibre-Purkinje cell synaptic responses. J Physiol 2016; 594:915-36. [PMID: 26627919 DOI: 10.1113/jp271623] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/01/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS At the parallel fibre-Purkinje cell glutamatergic synapse, little or no Ca(2+) entry takes place through postsynaptic neurotransmitter receptors, although postsynaptic calcium increases are clearly involved in the synaptic plasticity. Postsynaptic voltage-gated Ca(2+) channels therefore constitute the sole rapid postsynaptic Ca(2+) signalling mechanism, making it essential to understand how they contribute to the synaptic signalling. Using a selective T-type calcium channel antagonist, we describe a T-type component of the EPSC that is activated by the AMPA receptor-mediated depolarization of the spine and thus will contribute to the local calcium dynamics. This component can amount up to 20% of the EPSC, and this fraction is maintained even at the high frequencies sometimes encountered in sensory processing. Modelling based on our biophysical characterization of T-type calcium channels in Purkinje cells suggests that the brief spine EPSCs cause the activated T-type channels to deactivate rather than inactivate, enabling repetitive activation. ABSTRACT In the cerebellum, sensory information is conveyed to Purkinje cells (PC) via the granule cell/parallel fibre (PF) pathway. Plasticity at the PF-PC synapse is considered to be a mechanism of information storage in motor learning. The induction of synaptic plasticity in the cerebellum and elsewhere usually involves intracellular Ca(2+) signals. Unusually, postsynaptic Ca(2+) signalling in PF-PC spines does not involve ionotropic glutamatergic receptors because postsynaptic NMDA receptors are absent and the AMPA receptors are Ca(2+) -impermeable; postsynaptic voltage-gated Ca(2+) channels therefore constitute the sole rapid Ca(2+) signalling mechanism. Low-threshold activated T-type calcium channels are present at the synapse, although their contribution to PF-PC synaptic responses is unknown. Taking advantage of 3,5-dichloro-N-[1-(2,2-dimethyl-tetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide, a selective T-type channel antagonist, we show in the mouse that inhibition of these channels reduces PF-PC excitatory postsynaptic currents and excitatory postsynaptic potentials by 15-20%. This contribution was preserved during sparse input and repetitive activity. We characterized the biophysical properties of native T-type channels in young animals and modelled their activation during simulated dendritic excitatory postsynaptic potential waveforms. The comparison of modelled and observed synaptic responses suggests that T-type channels only activate in spines that are strongly depolarized by their synaptic input, a process requiring a high spine neck resistance. This brief and local activation ensures that T-type channels rapidly deactivate, thereby limiting inactivation during repetitive synaptic activity. T-type channels are therefore ideally situated to provide synaptic Ca(2+) entry at PF-PC spines.
Collapse
Affiliation(s)
- Romain Ly
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS UMR 8197 and INSERM U1024, Paris, France
| | - Guy Bouvier
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS UMR 8197 and INSERM U1024, Paris, France
| | - German Szapiro
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS UMR 8197 and INSERM U1024, Paris, France
| | - Haydn M Prosser
- GlaxoSmithKline Pharmaceuticals, New Frontiers Science Park, Third Avenue, Harlow, UK., Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Andrew D Randall
- GlaxoSmithKline Pharmaceuticals, New Frontiers Science Park, Third Avenue, Harlow, UK.,School of Physiology and Pharmacology, Medical Sciences Building, University of Bristol, Bristol, UK
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Philippe Isope
- INCI, CNRS UPR 3212, Centre de Neurochimie, Strasbourg, France
| | - Boris Barbour
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS UMR 8197 and INSERM U1024, Paris, France
| | - Anne Feltz
- Ecole Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS UMR 8197 and INSERM U1024, Paris, France
| |
Collapse
|
35
|
Inhibition promotes long-term potentiation at cerebellar excitatory synapses. Sci Rep 2016; 6:33561. [PMID: 27641070 PMCID: PMC5027531 DOI: 10.1038/srep33561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/30/2016] [Indexed: 12/18/2022] Open
Abstract
The ability of the cerebellar cortex to learn from experience ensures the accuracy of movements and reflex adaptation, processes which require long-term plasticity at granule cell (GC) to Purkinje neuron (PN) excitatory synapses. PNs also receive GABAergic inhibitory inputs via GCs activation of interneurons; despite the involvement of inhibition in motor learning, its role in long-term plasticity is poorly characterized. Here we reveal a functional coupling between ionotropic GABAA receptors and low threshold CaV3 calcium channels in PNs that sustains calcium influx and promotes long-term potentiation (LTP) at GC to PN synapses. High frequency stimulation induces LTP at GC to PN synapses and CaV3-mediated calcium influx provided that inhibition is intact; LTP is mGluR1, intracellular calcium store and CaV3 dependent. LTP is impaired in CaV3.1 knockout mice but it is nevertheless recovered by strengthening inhibitory transmission onto PNs; promoting a stronger hyperpolarization via GABAA receptor activation leads to an enhanced availability of an alternative Purkinje-expressed CaV3 isoform compensating for the lack of CaV3.1 and restoring LTP. Accordingly, a stronger hyperpolarization also restores CaV3-mediated calcium influx in PNs from CaV3.1 knockout mice. We conclude that by favoring CaV3 channels availability inhibition promotes LTP at cerebellar excitatory synapses.
Collapse
|
36
|
Aguado C, García-Madrona S, Gil-Minguez M, Luján R. Ontogenic Changes and Differential Localization of T-type Ca(2+) Channel Subunits Cav3.1 and Cav3.2 in Mouse Hippocampus and Cerebellum. Front Neuroanat 2016; 10:83. [PMID: 27616982 PMCID: PMC4999439 DOI: 10.3389/fnana.2016.00083] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/11/2016] [Indexed: 12/31/2022] Open
Abstract
T-type calcium (Ca(2+)) channels play a central role in regulating membrane excitability in the brain. Although the contributions of T-type current to neuron output is often proposed to reflect a differential distribution of T-type channel subtypes to somato-dendritic compartments, their precise subcellular distributions in central neurons are not fully determined. Using histoblot and high-resolution immunoelectron microscopic techniques, we have investigated the expression, regional distribution and subcellular localization of T-type Cav3.1 and Cav3.2 channel subunits in the adult brain, as well as the ontogeny of expression during postnatal development. Histoblot analysis showed that Cav3.1 and Cav3.2 proteins were widely expressed in the brain, with mostly non-overlapping patterns. Cav3.1 showed the highest expression level in the molecular layer (ml) of the cerebellum (Cb), and Cav3.2 in the hippocampus (Hp) and the ml of Cb. During development, levels of Cav3.1 and Cav3.2 increased with age, although there were marked region- and developmental stage-specific differences in their expression. At the cellular and subcellular level, immunoelectron microscopy showed that labeling for Cav3.1 was present in somato-dendritic domains of hippocampal interneurons and Purkinje cells (PCs), while Cav3.2 was present in somato-dendritic domains of CA1 pyramidal cells, hippocampal interneurons and PCs. Most of the immunoparticles for Cav3.1 and Cav3.2 were either associated with the plasma membrane or the intracellular membranes, with notable differences depending on the compartment. Thus, Cav3.1 was mainly located in the plasma membrane of interneurons, whereas Cav3.2 was mainly located in the plasma membrane of dendritic spines and had a major intracellular distribution in dendritic shafts. In PCs, Cav3.1 and Cav3.2 showed similar distribution patterns. In addition to its main postsynaptic distribution, Cav3.2 but not Cav3.1 was also detected in axon terminals establishing excitatory synapses. These results shed new light on the subcellular localization of T-type channel subunits and provide evidence for the non-uniform distribution of Cav3.1 and Cav3.2 subunits over the plasma membrane of central neurons, which may account for the functional heterogeneity of T-type mediated current.
Collapse
Affiliation(s)
- Carolina Aguado
- Synaptic Structure Laboratory, Department Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| | - Sebastián García-Madrona
- Synaptic Structure Laboratory, Department Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| | - Mercedes Gil-Minguez
- Synaptic Structure Laboratory, Department Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| | - Rafael Luján
- Synaptic Structure Laboratory, Department Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| |
Collapse
|
37
|
Matsumoto-Makidono Y, Nakayama H, Yamasaki M, Miyazaki T, Kobayashi K, Watanabe M, Kano M, Sakimura K, Hashimoto K. Ionic Basis for Membrane Potential Resonance in Neurons of the Inferior Olive. Cell Rep 2016; 16:994-1004. [PMID: 27425615 DOI: 10.1016/j.celrep.2016.06.053] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/04/2016] [Accepted: 06/12/2016] [Indexed: 11/25/2022] Open
Abstract
Some neurons have the ability to enhance output voltage to input current with a preferred frequency, which is called resonance. Resonance is thought to be a basis for membrane potential oscillation. Although ion channels responsible for resonance have been reported, the precise mechanisms by which these channels work remain poorly understood. We have found that resonance is reduced but clearly present in the inferior olivary neurons of Cav3.1 T-type voltage-dependent Ca(2+) channel knockout (KO) mice. The activation of Cav3.1 channels is strongly membrane potential dependent, but less frequency dependent. Residual resonance in Cav3.1 KO mice is abolished by a hyper-polarization-activated cyclic nucleotide-gated (HCN) channel blocker, ZD7288, and is partially suppressed by voltage-dependent K(+) channel blockers. Resonance is inhibited by ZD7288 in wild-type mice and impaired in HCN1 KO mice, suggesting that the HCN1 channel is essential for resonance. The ZD7288-sensitive current is nearly sinusoidal and strongly frequency dependent. These results suggest that Cav3.1 and HCN1 channels act as amplifying and resonating conductances, respectively.
Collapse
Affiliation(s)
- Yoshiko Matsumoto-Makidono
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8551, Japan
| | - Hisako Nakayama
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8551, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Taisuke Miyazaki
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo 113-0033, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8551, Japan.
| |
Collapse
|
38
|
Chen E, Paré JF, Wichmann T, Smith Y. Sub-synaptic localization of Ca v3.1 T-type calcium channels in the thalamus of normal and parkinsonian monkeys. Brain Struct Funct 2016; 222:735-748. [PMID: 27255751 DOI: 10.1007/s00429-016-1242-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/20/2016] [Indexed: 11/25/2022]
Abstract
T-type calcium channels (Cav3) are key mediators of thalamic bursting activity, but also regulate single cells excitability, dendritic integration, synaptic strength and transmitter release. These functions are strongly influenced by the subcellular and subsynaptic localization of Cav3 channels along the somatodendritic domain of thalamic cells. In Parkinson's disease, T-type calcium channels dysfunction in the basal ganglia-receiving thalamic nuclei likely contributes to pathological thalamic bursting activity. In this study, we analyzed the cellular, subcellular, and subsynaptic localization of the Cav3.1 channel in the ventral anterior (VA) and centromedian/parafascicular (CM/Pf) thalamic nuclei, the main thalamic targets of basal ganglia output, in normal and parkinsonian monkeys. All thalamic nuclei displayed strong Cav3.1 neuropil immunoreactivity, although the intensity of immunolabeling in CM/Pf was significantly lower than in VA. Ultrastructurally, 70-80 % of the Cav3.1-immunoreactive structures were dendritic shafts. Using immunogold labeling, Cav3.1 was commonly found perisynaptic to asymmetric and symmetric axo-dendritic synapses, suggesting a role of Cav3.1 in regulating excitatory and inhibitory neurotransmission. Significant labeling was also found at non-synaptic sites along the plasma membrane of thalamic neurons. There was no difference in the overall pattern and intensity of immunostaining between normal and parkinsonian monkeys, suggesting that the increased rebound bursting in the parkinsonian state is not driven by changes in Cav3.1 expression. Thus, T-type calcium channels are located to subserve neuronal bursting, but also regulate glutamatergic and non-glutamatergic transmission along the whole somatodendritic domain of basal ganglia-receiving neurons of the primate thalamus.
Collapse
Affiliation(s)
- Erdong Chen
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, 30322, USA
| | - Jean-Francois Paré
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, 30322, USA
| | - Thomas Wichmann
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA. .,Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, GA, 30322, USA. .,Department of Neurology, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
39
|
Medial septal GABAergic projection neurons promote object exploration behavior and type 2 theta rhythm. Proc Natl Acad Sci U S A 2016; 113:6550-5. [PMID: 27208094 DOI: 10.1073/pnas.1605019113] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Exploratory drive is one of the most fundamental emotions, of all organisms, that are evoked by novelty stimulation. Exploratory behavior plays a fundamental role in motivation, learning, and well-being of organisms. Diverse exploratory behaviors have been described, although their heterogeneity is not certain because of the lack of solid experimental evidence for their distinction. Here we present results demonstrating that different neural mechanisms underlie different exploratory behaviors. Localized Cav3.1 knockdown in the medial septum (MS) selectively enhanced object exploration, whereas the null mutant (KO) mice showed enhanced-object exploration as well as open-field exploration. In MS knockdown mice, only type 2 hippocampal theta rhythm was enhanced, whereas both type 1 and type 2 theta rhythm were enhanced in KO mice. This selective effect was accompanied by markedly increased excitability of septo-hippocampal GABAergic projection neurons in the MS lacking T-type Ca(2+) channels. Furthermore, optogenetic activation of the septo-hippocampal GABAergic pathway in WT mice also selectively enhanced object exploration behavior and type 2 theta rhythm, whereas inhibition of the same pathway decreased the behavior and the rhythm. These findings define object exploration distinguished from open-field exploration and reveal a critical role of T-type Ca(2+) channels in the medial septal GABAergic projection neurons in this behavior.
Collapse
|
40
|
Karimzadeh F, Modarres Mousavi SM, Ghadiri T, Jafarian M, Soleimani M, Sadeghi SM, Mesgari M, Joghataei MT, Gorji A. The Modulatory Effect of Metabotropic Glutamate Receptor Type-1α on Spike-Wave Discharges in WAG/Rij Rats. Mol Neurobiol 2016; 54:846-854. [PMID: 26780454 DOI: 10.1007/s12035-016-9692-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/05/2016] [Indexed: 02/06/2023]
Abstract
Modulatory function of metabotropic glutamate type 1 (mGlu1) receptors plays a crucial role in the pathophysiology of some neurological disorders, including schizophrenia and epilepsy. In this study, the expression of mGlu1α receptors in the thalamic nuclei was assessed during development of absence seizures in the WAG/Rij rats, a valid genetic animal model of absence epilepsy. In addition, the effect of pharmacological modulation of mGlu1α receptors in the laterodorsal (LD) nucleus of the thalamus on the characteristic features of bioelectrical brain activities in the WAG/Rij rats was assessed. The expression of mGlu1α receptors in the LD was assessed in four experimental groups of both WAG/Rij and Wistar rats with 2 and 6 months of age. Agonist and antagonist of mGlu1α receptors were infused in LD in the six months old WAG/Rij (epileptic) rats. The protein level of mGlu1α receptors in the thalamus of the 6-month-old WAG/Rij rats was lower than non-epileptic animals. In addition, the distribution of mGlu1α receptors in different thalamic nuclei was lower in the 6-month-old WAG/Rij compared to age-matched Wistar rats. The gene expression of mGlu1α receptor was also significantly lower in 6-month-old WAG/Rij rats in the LD compared to other animal groups. The microinjection of mGlu1α receptors agonist and antagonist in the LD reduced the duration of spike-wave discharges (SWDs) and increased the amplitude and duration of SWDs, respectively, in 6-month-old WAG/Rij rats. The alterations of mGlu1α receptors expression in the thalamus of epileptic WAG/Rij rats as well as its modulatory effects in the generation of SWDs suggest the potential of mGlu1 receptors as a therapeutic target in absence epilepsy.
Collapse
Affiliation(s)
- Fariba Karimzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Tahereh Ghadiri
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Maryam Jafarian
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shahin Mohammad Sadeghi
- Department of Plastic and Reconstructive Surgery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Mesgari
- Klinik und Poliklinik für Neurochirurgie, Westfälische Wilhelms-Universität Münster, Münster, Germany.,Klinik und Poliklinik für Neurologie, Westfälische Wilhelms-Universität Münster, Münster, Germany.,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran. .,Klinik und Poliklinik für Neurochirurgie, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Klinik und Poliklinik für Neurologie, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| |
Collapse
|
41
|
Morino H, Matsuda Y, Muguruma K, Miyamoto R, Ohsawa R, Ohtake T, Otobe R, Watanabe M, Maruyama H, Hashimoto K, Kawakami H. A mutation in the low voltage-gated calcium channel CACNA1G alters the physiological properties of the channel, causing spinocerebellar ataxia. Mol Brain 2015; 8:89. [PMID: 26715324 PMCID: PMC4693440 DOI: 10.1186/s13041-015-0180-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 12/21/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Spinocerebellar ataxia (SCA) is a genetically heterogeneous disease. To date, 36 dominantly inherited loci have been reported, and 31 causative genes have been identified. RESULTS In this study, we analyzed a Japanese family with autosomal dominant SCA using linkage analysis and exome sequencing, and identified CACNA1G, which encodes the calcium channel CaV3.1, as a new causative gene. The same mutation was also found in another family with SCA. Although most patients exhibited the pure form of cerebellar ataxia, two patients showed prominent resting tremor in addition to ataxia. CaV3.1 is classified as a low-threshold voltage-dependent calcium channel (T-type) and is expressed abundantly in the central nervous system, including the cerebellum. The mutation p.Arg1715His, identified in this study, was found to be located at S4 of repeat IV, the voltage sensor of the CaV3.1. Electrophysiological analyses revealed that the membrane potential dependency of the mutant CaV3.1 transfected into HEK293T cells shifted toward a positive potential. We established induced pluripotent stem cells (iPSCs) from fibroblasts of the patient, and to our knowledge, this is the first report of successful differentiation from the patient-derived iPSCs into Purkinje cells. There was no significant difference in the differentiation status between control- and patient-derived iPSCs. CONCLUSIONS To date, several channel genes have been reported as causative genes for SCA. Our findings provide important insights into the pathogenesis of SCA as a channelopathy.
Collapse
Affiliation(s)
- Hiroyuki Morino
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Yukiko Matsuda
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Keiko Muguruma
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe, Japan.
| | - Ryosuke Miyamoto
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Ryosuke Ohsawa
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Toshiyuki Ohtake
- Department of Neurology, Tokyo Metropolitan Health and Medical Treatment Corporation Ebara Hospital, Tokyo, Japan.
| | - Reiko Otobe
- Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima, Japan.
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience & Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Hideshi Kawakami
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| |
Collapse
|
42
|
Powell KL, Cain SM, Snutch TP, O'Brien TJ. Low threshold T-type calcium channels as targets for novel epilepsy treatments. Br J Clin Pharmacol 2015; 77:729-39. [PMID: 23834404 DOI: 10.1111/bcp.12205] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 07/02/2013] [Indexed: 12/21/2022] Open
Abstract
Low voltage-activated T-type calcium channels were originally cloned in the 1990s and much research has since focused on identifying the physiological roles of these channels in health and disease states. T-type calcium channels are expressed widely throughout the brain and peripheral tissues, and thus have been proposed as therapeutic targets for a variety of diseases such as epilepsy, insomnia, pain, cancer and hypertension. This review discusses the literature concerning the role of T-type calcium channels in physiological and pathological processes related to epilepsy. T-type calcium channels have been implicated in pathology of both the genetic and acquired epilepsies and several anti-epileptic drugs (AEDs) in clinical use are known to suppress seizures via inhibition of T-type calcium channels. Despite the fact that more than 15 new AEDs have become clinically available over the past 20 years at least 30% of epilepsy patients still fail to achieve seizure control, and many patients experience unwanted side effects. Furthermore there are no treatments that prevent the development of epilepsy or mitigate the epileptic state once established. Therefore there is an urgent need for the development of new AEDs that are effective in patients with drug resistant epilepsy, are anti-epileptogenic and are better tolerated. We also review the mechanisms of action of the current AEDs with known effects on T-type calcium channels and discuss novel compounds that are being investigated as new treatments for epilepsy.
Collapse
Affiliation(s)
- Kim L Powell
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
43
|
Marcaggi P. Cerebellar Endocannabinoids: Retrograde Signaling from Purkinje Cells. THE CEREBELLUM 2014; 14:341-53. [DOI: 10.1007/s12311-014-0629-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
44
|
Otsu Y, Marcaggi P, Feltz A, Isope P, Kollo M, Nusser Z, Mathieu B, Kano M, Tsujita M, Sakimura K, Dieudonné S. Activity-dependent gating of calcium spikes by A-type K+ channels controls climbing fiber signaling in Purkinje cell dendrites. Neuron 2014; 84:137-151. [PMID: 25220810 PMCID: PMC4183427 DOI: 10.1016/j.neuron.2014.08.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2014] [Indexed: 12/01/2022]
Abstract
In cerebellar Purkinje cell dendrites, heterosynaptic calcium signaling induced by the proximal climbing fiber (CF) input controls plasticity at distal parallel fiber (PF) synapses. The substrate and regulation of this long-range dendritic calcium signaling are poorly understood. Using high-speed calcium imaging, we examine the role of active dendritic conductances. Under basal conditions, CF stimulation evokes T-type calcium signaling displaying sharp proximodistal decrement. Combined mGluR1 receptor activation and depolarization, two activity-dependent signals, unlock P/Q calcium spikes initiation and propagation, mediating efficient CF signaling at distal sites. These spikes are initiated in proximal smooth dendrites, independently from somatic sodium action potentials, and evoke high-frequency bursts of all-or-none fast-rising calcium transients in PF spines. Gradual calcium spike burst unlocking arises from increasing inactivation of mGluR1-modulated low-threshold A-type potassium channels located in distal dendrites. Evidence for graded activity-dependent CF calcium signaling at PF synapses refines current views on cerebellar supervised learning rules.
Collapse
Affiliation(s)
- Yo Otsu
- Inhibitory Transmission Team, IBENS, CNRS UMR UMR8197, INSERM U1024, Ecole Normale Supérieure, 75005 Paris, France
| | - Païkan Marcaggi
- Inhibitory Transmission Team, IBENS, CNRS UMR UMR8197, INSERM U1024, Ecole Normale Supérieure, 75005 Paris, France
| | - Anne Feltz
- Cerebellum Group, IBENS, CNRS UMR UMR8197, INSERM U1024, Ecole Normale Supérieure, 75005 Paris, France
| | - Philippe Isope
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, 67000-Strasbourg, France
| | - Mihaly Kollo
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine of the Hungarian Academy of Sciences, 1083 Budapest, Hungary
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine of the Hungarian Academy of Sciences, 1083 Budapest, Hungary
| | - Benjamin Mathieu
- Imaging Facility, IBENS, CNRS UMR 8197, INSERM U1024, Ecole Normale Supérieure, 75005 Paris, France
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Mika Tsujita
- Center for Transdisciplinary Research, Niigata University, Niigata 950-2181, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Stéphane Dieudonné
- Inhibitory Transmission Team, IBENS, CNRS UMR UMR8197, INSERM U1024, Ecole Normale Supérieure, 75005 Paris, France.
| |
Collapse
|
45
|
Subramaniyam S, Solinas S, Perin P, Locatelli F, Masetto S, D'Angelo E. Computational modeling predicts the ionic mechanism of late-onset responses in unipolar brush cells. Front Cell Neurosci 2014; 8:237. [PMID: 25191224 PMCID: PMC4138490 DOI: 10.3389/fncel.2014.00237] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/27/2014] [Indexed: 11/29/2022] Open
Abstract
Unipolar Brush Cells (UBCs) have been suggested to play a critical role in cerebellar functioning, yet the corresponding cellular mechanisms remain poorly understood. UBCs have recently been reported to generate, in addition to early-onset glutamate receptor-dependent synaptic responses, a late-onset response (LOR) composed of a slow depolarizing ramp followed by a spike burst (Locatelli et al., 2013). The LOR activates as a consequence of synaptic activity and involves an intracellular cascade modulating H- and TRP-current gating. In order to assess the LOR mechanisms, we have developed a UBC multi-compartmental model (including soma, dendrite, initial segment, and axon) incorporating biologically realistic representations of ionic currents and a cytoplasmic coupling mechanism regulating TRP and H channel gating. The model finely reproduced UBC responses to current injection, including a burst triggered by a low-threshold spike (LTS) sustained by CaLVA currents, a persistent discharge sustained by CaHVA currents, and a rebound burst following hyperpolarization sustained by H- and CaLVA-currents. Moreover, the model predicted that H- and TRP-current regulation was necessary and sufficient to generate the LOR and its dependence on the intensity and duration of mossy fiber activity. Therefore, the model showed that, using a basic set of ionic channels, UBCs generate a rich repertoire of bursts, which could effectively implement tunable delay-lines in the local microcircuit.
Collapse
Affiliation(s)
- Sathyaa Subramaniyam
- Neurophysiology Unit, Department of Brain and Behavioral Science, University of Pavia Pavia, Italy ; Consorzio Interuniversitario per le Scienze Fisiche della Materia (CNISM) Pavia, Italy
| | - Sergio Solinas
- Neurophysiology Unit, Brain Connectivity Center, Istituto Neurologico IRCCS C. Mondino Pavia, Italy
| | - Paola Perin
- Neurophysiology Unit, Department of Brain and Behavioral Science, University of Pavia Pavia, Italy
| | - Francesca Locatelli
- Neurophysiology Unit, Department of Brain and Behavioral Science, University of Pavia Pavia, Italy
| | - Sergio Masetto
- Neurophysiology Unit, Department of Brain and Behavioral Science, University of Pavia Pavia, Italy
| | - Egidio D'Angelo
- Neurophysiology Unit, Department of Brain and Behavioral Science, University of Pavia Pavia, Italy ; Neurophysiology Unit, Brain Connectivity Center, Istituto Neurologico IRCCS C. Mondino Pavia, Italy
| |
Collapse
|
46
|
Chamberland S, Evstratova A, Tóth K. Interplay between synchronization of multivesicular release and recruitment of additional release sites support short-term facilitation at hippocampal mossy fiber to CA3 pyramidal cells synapses. J Neurosci 2014; 34:11032-47. [PMID: 25122902 PMCID: PMC6705252 DOI: 10.1523/jneurosci.0847-14.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/07/2014] [Accepted: 06/27/2014] [Indexed: 11/21/2022] Open
Abstract
Synaptic short-term plasticity is a key regulator of neuronal communication and is controlled via various mechanisms. A well established property of mossy fiber to CA3 pyramidal cell synapses is the extensive short-term facilitation during high-frequency bursts. We investigated the mechanisms governing facilitation using a combination of whole-cell electrophysiological recordings, electrical minimal stimulation, and random-access two-photon microscopy in acute mouse hippocampal slices. Two distinct presynaptic mechanisms were involved in short-term facilitation, with their relative contribution dependent on extracellular calcium concentration. The synchronization of multivesicular release was observed during trains of facilitating EPSCs recorded in 1.2 mM external Ca(2+) ([Ca(2+)]e). Indeed, covariance analysis revealed a gradual augmentation in quantal size during trains of EPSCs, and application of the low-affinity glutamate receptor antagonist γ-D-glutamylglycine showed an increase in cleft glutamate concentration during paired-pulse stimulation. Whereas synchronization of multivesicular release contributed to the facilitation in 1.2 mM [Ca(2+)]e, variance-mean analysis showed that recruitment of more release sites (N) was likely to account for the larger facilitation observed in 2.5 mM [Ca(2+)]e. Furthermore, this increase in N could be promoted by calcium microdomains of heterogeneous amplitudes observed in single mossy fiber boutons. Our findings suggest that the combination of multivesicular release and the recruitment of additional release sites act together to increase glutamate release during burst activity. This is supported by the compartmentalized spatial profile of calcium elevations in boutons and helps to expand the dynamic range of mossy fibers information transfer.
Collapse
Affiliation(s)
- Simon Chamberland
- Quebec Mental Health Institute, Department of Psychiatry and Neuroscience, Laval University, Quebec City, Quebec, Canada, G1J 2G3
| | - Alesya Evstratova
- Quebec Mental Health Institute, Department of Psychiatry and Neuroscience, Laval University, Quebec City, Quebec, Canada, G1J 2G3
| | - Katalin Tóth
- Quebec Mental Health Institute, Department of Psychiatry and Neuroscience, Laval University, Quebec City, Quebec, Canada, G1J 2G3
| |
Collapse
|
47
|
The role of canonical transient receptor potential channels in seizure and excitotoxicity. Cells 2014; 3:288-303. [PMID: 24722470 PMCID: PMC4092853 DOI: 10.3390/cells3020288] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/29/2014] [Accepted: 04/02/2014] [Indexed: 12/11/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are a family of polymodal cation channels with some degree of Ca2+ permeability. Although initially thought to be channels mediating store-operated Ca2+ influx, TRPC channels can be activated by stimulation of Gq-coupled G-protein coupled receptors, or by an increase in intracellular free Ca2+ concentration. Thus, activation of TRPC channels could be a common downstream event of many signaling pathways that contribute to seizure and excitotoxicity, such as N-methyl-D-aspartate (NMDA) receptor-mediated Ca2+ influx, or metabotropic glutamate receptor activation. Recent studies with genetic ablation of various TRPC family members have demonstrated that TRPC channels, in particular heteromeric TRPC1/4 channels and homomeric TRPC5 channels, play a critical role in both pilocarpine-induced acute seizures and neuronal cell death. However, exact underlying mechanisms remain to be fully elucidated, and selective TRPC modulators and antibodies with better specificity are urgently needed for future research.
Collapse
|
48
|
Engbers JDT, Anderson D, Zamponi GW, Turner RW. Signal processing by T-type calcium channel interactions in the cerebellum. Front Cell Neurosci 2013; 7:230. [PMID: 24348329 PMCID: PMC3841819 DOI: 10.3389/fncel.2013.00230] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/06/2013] [Indexed: 01/28/2023] Open
Abstract
T-type calcium channels of the Cav3 family are unique among voltage-gated calcium channels due to their low activation voltage, rapid inactivation, and small single channel conductance. These special properties allow Cav3 calcium channels to regulate neuronal processing in the subthreshold voltage range. Here, we review two different subthreshold ion channel interactions involving Cav3 channels and explore the ability of these interactions to expand the functional roles of Cav3 channels. In cerebellar Purkinje cells, Cav3 and intermediate conductance calcium-activated potassium (IKCa) channels form a novel complex which creates a low voltage-activated, transient outward current capable of suppressing temporal summation of excitatory postsynaptic potentials (EPSPs). In large diameter neurons of the deep cerebellar nuclei, Cav3-mediated calcium current (I T) and hyperpolarization-activated cation current (I H) are activated during trains of inhibitory postsynaptic potentials. These currents have distinct, and yet synergistic, roles in the subthreshold domain with I T generating a rebound burst and I H controlling first spike latency and rebound spike precision. However, by shortening the membrane time constant the membrane returns towards resting value at a faster rate, allowing I H to increase the efficacy of I T and increase the range of burst frequencies that can be generated. The net effect of Cav3 channels thus depends on the channels with which they are paired. When expressed in a complex with a KCa channel, Cav3 channels reduce excitability when processing excitatory inputs. If functionally coupled with an HCN channel, the depolarizing effect of Cav3 channels is accentuated, allowing for efficient inversion of inhibitory inputs to generate a rebound burst output. Therefore, signal processing relies not only on the activity of individual subtypes of channels but also on complex interactions between ion channels whether based on a physical complex or by indirect effects on membrane properties.
Collapse
Affiliation(s)
- Jordan D. T. Engbers
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of CalgaryCalgary, Canada
| | - Dustin Anderson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of CalgaryCalgary, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of CalgaryCalgary, Canada
| | - Ray W. Turner
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of CalgaryCalgary, Canada
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of CalgaryCalgary, Canada
| |
Collapse
|
49
|
T-type channel blockade impairs long-term potentiation at the parallel fiber-Purkinje cell synapse and cerebellar learning. Proc Natl Acad Sci U S A 2013; 110:20302-7. [PMID: 24277825 DOI: 10.1073/pnas.1311686110] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
CaV3.1 T-type channels are abundant at the cerebellar synapse between parallel fibers and Purkinje cells where they contribute to synaptic depolarization. So far, no specific physiological function has been attributed to these channels neither as charge carriers nor more specifically as Ca(2+) carriers. Here we analyze their incidence on synaptic plasticity, motor behavior, and cerebellar motor learning, comparing WT animals and mice where T-type channel function has been abolished either by gene deletion or by acute pharmacological blockade. At the cellular level, we show that CaV3.1 channels are required for long-term potentiation at parallel fiber-Purkinje cell synapses. Moreover, basal simple spike discharge of the Purkinje cell in KO mice is modified. Acute or chronic T-type current blockade results in impaired motor performance in particular when a good body balance is required. Because motor behavior integrates reflexes and past memories of learned behavior, this suggests impaired learning. Indeed, subjecting the KO mice to a vestibulo-ocular reflex phase reversal test reveals impaired cerebellum-dependent motor learning. These data identify a role of low-voltage activated calcium channels in synaptic plasticity and establish a role for CaV3.1 channels in cerebellar learning.
Collapse
|
50
|
The plasma membrane Ca2+-ATPase2 (PMCA2) is involved in the regulation of Purkinje cell dendritic growth in cerebellar organotypic slice cultures. Neural Plast 2013; 2013:321685. [PMID: 24288624 PMCID: PMC3830849 DOI: 10.1155/2013/321685] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/30/2013] [Accepted: 09/03/2013] [Indexed: 11/17/2022] Open
Abstract
Purkinje cells are the principal neurons of the cerebellar cortex and have an extensive and elaborate dendritic tree. Chronic activation of type I metabotropic glutamate receptors inhibits Purkinje cell dendritic growth in organotypic cerebellar slice cultures. This effect is mediated by calcium influx through P/Q-type and T-type Ca2+ channels. We have now studied the role of the plasma membrane Ca2+-ATPase2 (PMCA2), a major calcium extrusion pump, for Purkinje cell dendritic development. We found that PMCA2 is strongly expressed in the plasma membrane and dendritic spines of Purkinje cells in organotypic slice cultures compatible with a role for controlling the local dendritic calcium equilibrium. Inhibition of PMCA2 activity by carboxyeosin resulted in a moderate reduction of Purkinje cell dendritic tree size indicating that the extrusion of calcium by PMCA2 is important for maintaining the dendritic calcium concentration and controlling dendritic growth. When inhibition of PMCA2 was combined with stimulation of type I metabotropic glutamate receptors, it partially rescued dendritic morphology. This protection can be explained by a compensatory inactivation of voltage-gated calcium channels in Purkinje cells after PMCA2 inhibition. Our results demonstrate that PMCA2 activity is an important regulator of the dendritic calcium equilibrium controlling Purkinje cell dendritic growth.
Collapse
|