1
|
Schloetter M, Maret GU, Kleineidam CJ. Annihilation of action potentials induces electrical coupling between neurons. eLife 2025; 12:RP88335. [PMID: 40183775 PMCID: PMC11970907 DOI: 10.7554/elife.88335] [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: 04/05/2025] Open
Abstract
Neurons generate and propagate electrical pulses called action potentials which annihilate on arrival at the axon terminal. We measure the extracellular electric field generated by propagating and annihilating action potentials and find that on annihilation, action potentials expel a local discharge. The discharge at the axon terminal generates an inhomogeneous electric field that immediately influences target neurons and thus provokes ephaptic coupling. Our measurements are quantitatively verified by a powerful analytical model which reveals excitation and inhibition in target neurons, depending on position and morphology of the source-target arrangement. Our model is in full agreement with experimental findings on ephaptic coupling at the well-studied Basket cell-Purkinje cell synapse. It is able to predict ephaptic coupling for any other synaptic geometry as illustrated by a few examples.
Collapse
Affiliation(s)
- Moritz Schloetter
- Department of Physics, University of KonstanzKonstanzGermany
- Neurobiology, Department of Biology, University of KonstanzKonstanzGermany
| | - Georg U Maret
- Department of Physics, University of KonstanzKonstanzGermany
| | | |
Collapse
|
2
|
Han X, Lu X, Li PH, Wang S, Schalek R, Meirovitch Y, Lin Z, Adhinarta J, Murray KD, MacNiven LM, Berger DR, Wu Y, Fang T, Meral ES, Asraf S, Ploegh H, Pfister H, Wei D, Jain V, Trimmer JS, Lichtman JW. Multiplexed volumetric CLEM enabled by scFvs provides insights into the cytology of cerebellar cortex. Nat Commun 2024; 15:6648. [PMID: 39103318 PMCID: PMC11300613 DOI: 10.1038/s41467-024-50411-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Mapping neuronal networks is a central focus in neuroscience. While volume electron microscopy (vEM) can reveal the fine structure of neuronal networks (connectomics), it does not provide molecular information to identify cell types or functions. We developed an approach that uses fluorescent single-chain variable fragments (scFvs) to perform multiplexed detergent-free immunolabeling and volumetric-correlated-light-and-electron-microscopy on the same sample. We generated eight fluorescent scFvs targeting brain markers. Six fluorescent probes were imaged in the cerebellum of a female mouse, using confocal microscopy with spectral unmixing, followed by vEM of the same sample. The results provide excellent ultrastructure superimposed with multiple fluorescence channels. Using this approach, we documented a poorly described cell type, two types of mossy fiber terminals, and the subcellular localization of one type of ion channel. Because scFvs can be derived from existing monoclonal antibodies, hundreds of such probes can be generated to enable molecular overlays for connectomic studies.
Collapse
Affiliation(s)
- Xiaomeng Han
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| | - Xiaotang Lu
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| | | | - Shuohong Wang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Richard Schalek
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Yaron Meirovitch
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Zudi Lin
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jason Adhinarta
- Computer Science Department, Boston College, Chestnut Hill, MA, USA
| | - Karl D Murray
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA, USA
| | - Leah M MacNiven
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA, USA
| | - Daniel R Berger
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Yuelong Wu
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Tao Fang
- Program of Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | - Shadnan Asraf
- School of Public Health, University of Massachusetts Amherst, Amherst, MA, USA
| | - Hidde Ploegh
- Program of Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Hanspeter Pfister
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA, USA
| | - Donglai Wei
- Computer Science Department, Boston College, Chestnut Hill, MA, USA
| | | | - James S Trimmer
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA, USA
| | - Jeff W Lichtman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
3
|
Ogawa Y, Lim BC, George S, Oses-Prieto JA, Rasband JM, Eshed-Eisenbach Y, Hamdan H, Nair S, Boato F, Peles E, Burlingame AL, Van Aelst L, Rasband MN. Antibody-directed extracellular proximity biotinylation reveals that Contactin-1 regulates axo-axonic innervation of axon initial segments. Nat Commun 2023; 14:6797. [PMID: 37884508 PMCID: PMC10603070 DOI: 10.1038/s41467-023-42273-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Axon initial segment (AIS) cell surface proteins mediate key biological processes in neurons including action potential initiation and axo-axonic synapse formation. However, few AIS cell surface proteins have been identified. Here, we use antibody-directed proximity biotinylation to define the cell surface proteins in close proximity to the AIS cell adhesion molecule Neurofascin. To determine the distributions of the identified proteins, we use CRISPR-mediated genome editing for insertion of epitope tags in the endogenous proteins. We identify Contactin-1 (Cntn1) as an AIS cell surface protein. Cntn1 is enriched at the AIS through interactions with Neurofascin and NrCAM. We further show that Cntn1 contributes to assembly of the AIS extracellular matrix, and regulates AIS axo-axonic innervation by inhibitory basket cells in the cerebellum and inhibitory chandelier cells in the cortex.
Collapse
Affiliation(s)
- Yuki Ogawa
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Brian C Lim
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shanu George
- Division of Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Joshua M Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hamdan Hamdan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Physiology and Immunology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Supna Nair
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Francesco Boato
- Division of Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Linda Van Aelst
- Division of Neuroscience, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Han X, Lu X, Li PH, Wang S, Schalek R, Meirovitch Y, Lin Z, Adhinarta J, Berger D, Wu Y, Fang T, Meral ES, Asraf S, Ploegh H, Pfister H, Wei D, Jain V, Trimmer JS, Lichtman JW. Multiplexed volumetric CLEM enabled by antibody derivatives provides new insights into the cytology of the mouse cerebellar cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.20.540091. [PMID: 37292964 PMCID: PMC10245788 DOI: 10.1101/2023.05.20.540091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mapping neuronal networks that underlie behavior has become a central focus in neuroscience. While serial section electron microscopy (ssEM) can reveal the fine structure of neuronal networks (connectomics), it does not provide the molecular information that helps identify cell types or their functional properties. Volumetric correlated light and electron microscopy (vCLEM) combines ssEM and volumetric fluorescence microscopy to incorporate molecular labeling into ssEM datasets. We developed an approach that uses small fluorescent single-chain variable fragment (scFv) immuno-probes to perform multiplexed detergent-free immuno-labeling and ssEM on the same samples. We generated eight such fluorescent scFvs that targeted useful markers for brain studies (green fluorescent protein, glial fibrillary acidic protein, calbindin, parvalbumin, voltage-gated potassium channel subfamily A member 2, vesicular glutamate transporter 1, postsynaptic density protein 95, and neuropeptide Y). To test the vCLEM approach, six different fluorescent probes were imaged in a sample of the cortex of a cerebellar lobule (Crus 1), using confocal microscopy with spectral unmixing, followed by ssEM imaging of the same sample. The results show excellent ultrastructure with superimposition of the multiple fluorescence channels. Using this approach we could document a poorly described cell type in the cerebellum, two types of mossy fiber terminals, and the subcellular localization of one type of ion channel. Because scFvs can be derived from existing monoclonal antibodies, hundreds of such probes can be generated to enable molecular overlays for connectomic studies.
Collapse
|
5
|
Martin HGS, Kullmann DM. Basket to Purkinje Cell Inhibitory Ephaptic Coupling Is Abolished in Episodic Ataxia Type 1. Cells 2023; 12:1382. [PMID: 37408217 PMCID: PMC10216961 DOI: 10.3390/cells12101382] [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: 03/15/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 07/07/2023] Open
Abstract
Dominantly inherited missense mutations of the KCNA1 gene, which encodes the KV1.1 potassium channel subunit, cause Episodic Ataxia type 1 (EA1). Although the cerebellar incoordination is thought to arise from abnormal Purkinje cell output, the underlying functional deficit remains unclear. Here we examine synaptic and non-synaptic inhibition of Purkinje cells by cerebellar basket cells in an adult mouse model of EA1. The synaptic function of basket cell terminals was unaffected, despite their intense enrichment for KV1.1-containing channels. In turn, the phase response curve quantifying the influence of basket cell input on Purkine cell output was maintained. However, ultra-fast non-synaptic ephaptic coupling, which occurs in the cerebellar 'pinceau' formation surrounding the axon initial segment of Purkinje cells, was profoundly reduced in EA1 mice in comparison with their wild type littermates. The altered temporal profile of basket cell inhibition of Purkinje cells underlines the importance of Kv1.1 channels for this form of signalling, and may contribute to the clinical phenotype of EA1.
Collapse
Affiliation(s)
| | - Dimitri M. Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK;
| |
Collapse
|
6
|
Kilisch M, Gere-Becker M, Wüstefeld L, Bonnas C, Crauel A, Mechmershausen M, Martens H, Götzke H, Opazo F, Frey S. Simple and Highly Efficient Detection of PSD95 Using a Nanobody and Its Recombinant Heavy-Chain Antibody Derivatives. Int J Mol Sci 2023; 24:ijms24087294. [PMID: 37108454 PMCID: PMC10138605 DOI: 10.3390/ijms24087294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The post-synaptic density protein 95 (PSD95) is a crucial scaffolding protein participating in the organization and regulation of synapses. PSD95 interacts with numerous molecules, including neurotransmitter receptors and ion channels. The functional dysregulation of PSD95 as well as its abundance and localization has been implicated with several neurological disorders, making it an attractive target for developing strategies able to monitor PSD95 accurately for diagnostics and therapeutics. This study characterizes a novel camelid single-domain antibody (nanobody) that binds strongly and with high specificity to rat, mouse, and human PSD95. This nanobody allows for more precise detection and quantification of PSD95 in various biological samples. We expect that the flexibility and unique performance of this thoroughly characterized affinity tool will help to further understand the role of PSD95 in normal and diseased neuronal synapses.
Collapse
Affiliation(s)
- Markus Kilisch
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Maja Gere-Becker
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Liane Wüstefeld
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Christel Bonnas
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Alexander Crauel
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Maja Mechmershausen
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Henrik Martens
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Hansjörg Götzke
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Felipe Opazo
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Steffen Frey
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| |
Collapse
|
7
|
Ogawa Y, Lim BC, George S, Oses-Prieto JA, Rasband JM, Eshed-Eisenbach Y, Nair S, Boato F, Peles E, Burlingame AL, Van Aelst L, Rasband MN. Antibody-directed extracellular proximity biotinylation reveals Contactin-1 regulates axo-axonic innervation of axon initial segments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531378. [PMID: 36945454 PMCID: PMC10028829 DOI: 10.1101/2023.03.06.531378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Axon initial segment (AIS) cell surface proteins mediate key biological processes in neurons including action potential initiation and axo-axonic synapse formation. However, few AIS cell surface proteins have been identified. Here, we used antibody-directed proximity biotinylation to define the cell surface proteins in close proximity to the AIS cell adhesion molecule Neurofascin. To determine the distributions of the identified proteins, we used CRISPR-mediated genome editing for insertion of epitope tags in the endogenous proteins. We found Contactin-1 (Cntn1) among the previously unknown AIS proteins we identified. Cntn1 is enriched at the AIS through interactions with Neurofascin and NrCAM. We further show that Cntn1 contributes to assembly of the AIS-extracellular matrix, and is required for AIS axo-axonic innervation by inhibitory basket cells in the cerebellum and inhibitory chandelier cells in the cortex.
Collapse
Affiliation(s)
- Yuki Ogawa
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| | - Brian C. Lim
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| | - Shanu George
- Cold Spring Harbor Laboratory, Division of Neuroscience, Cold Spring Harbor, NY, USA
| | - Juan A. Oses-Prieto
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA, USA
| | - Joshua M. Rasband
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| | - Yael Eshed-Eisenbach
- Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot, Israel
| | - Supna Nair
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA, USA
| | - Francesco Boato
- Cold Spring Harbor Laboratory, Division of Neuroscience, Cold Spring Harbor, NY, USA
| | - Elior Peles
- Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot, Israel
| | - Alma L. Burlingame
- University of California San Francisco, Department of Pharmaceutical Chemistry, San Francisco, CA, USA
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Division of Neuroscience, Cold Spring Harbor, NY, USA
| | - Matthew N. Rasband
- Baylor College of Medicine, Department of Neuroscience, Houston, TX, USA
| |
Collapse
|
8
|
Dziadkowiak E, Nowakowska-Kotas M, Budrewicz S, Koszewicz M. Pathology of Initial Axon Segments in Chronic Inflammatory Demyelinating Polyradiculoneuropathy and Related Disorders. Int J Mol Sci 2022; 23:13621. [PMID: 36362407 PMCID: PMC9658771 DOI: 10.3390/ijms232113621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 07/30/2023] Open
Abstract
The diagnosis of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is based on a combination of clinical, electrodiagnostic and laboratory features. The different entities of the disease include chronic immune sensory polyradiculopathy (CISP) and autoimmune nodopathies. It is debatable whether CIDP occurring in the course of other conditions, i.e., monoclonal IgG or IgA gammopathy, should be treated as a separate disease entity from idiopathic CIDP. This study aims to evaluate the molecular differences of the nodes of Ranvier and the initial axon segment (AIS) and juxtaparanode region (JXP) as the potential cause of phenotypic variation of CIDP while also seeking new pathomechanisms since JXP is sequestered behind the paranode and autoantibodies may not access the site easily. The authors initially present the structure of the different parts of the neuron and its functional significance, then discuss the problem of whether damage to the juxtaparanodal region, Schwann cells and axons could cause CIDP or if these damages should be separated as separate disease entities. In particular, AIS's importance for modulating neural excitability and carrying out transport along the axon is highlighted. The disclosure of specific pathomechanisms, including novel target antigens, in the heterogeneous CIDP syndrome is important for diagnosing and treating these patients.
Collapse
|
9
|
Masoli S, Rizza MF, Tognolina M, Prestori F, D’Angelo E. Computational models of neurotransmission at cerebellar synapses unveil the impact on network computation. Front Comput Neurosci 2022; 16:1006989. [PMID: 36387305 PMCID: PMC9649760 DOI: 10.3389/fncom.2022.1006989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
The neuroscientific field benefits from the conjoint evolution of experimental and computational techniques, allowing for the reconstruction and simulation of complex models of neurons and synapses. Chemical synapses are characterized by presynaptic vesicle cycling, neurotransmitter diffusion, and postsynaptic receptor activation, which eventually lead to postsynaptic currents and subsequent membrane potential changes. These mechanisms have been accurately modeled for different synapses and receptor types (AMPA, NMDA, and GABA) of the cerebellar cortical network, allowing simulation of their impact on computation. Of special relevance is short-term synaptic plasticity, which generates spatiotemporal filtering in local microcircuits and controls burst transmission and information flow through the network. Here, we present how data-driven computational models recapitulate the properties of neurotransmission at cerebellar synapses. The simulation of microcircuit models is starting to reveal how diverse synaptic mechanisms shape the spatiotemporal profiles of circuit activity and computation.
Collapse
Affiliation(s)
- Stefano Masoli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | | | | | - Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Brain Connectivity Center, Pavia, Italy
| |
Collapse
|
10
|
Lebenheim L, Booker SA, Derst C, Weiss T, Wagner F, Gruber C, Vida I, Zahm DS, Veh RW. A novel giant non-cholinergic striatal interneuron restricted to the ventrolateral striatum coexpresses Kv3.3 potassium channel, parvalbumin, and the vesicular GABA transporter. Mol Psychiatry 2022; 27:2315-2328. [PMID: 33190145 PMCID: PMC9126804 DOI: 10.1038/s41380-020-00948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The striatum is the main input structure of the basal ganglia. Distinct striatal subfields are involved in voluntary movement generation and cognitive and emotional tasks, but little is known about the morphological and molecular differences of striatal subregions. The ventrolateral subfield of the striatum (VLS) is the orofacial projection field of the sensorimotor cortex and is involved in the development of orofacial dyskinesias, involuntary chewing-like movements that often accompany long-term neuroleptic treatment. The biological basis for this particular vulnerability of the VLS is not known. Potassium channels are known to be strategically localized within the striatum. In search of possible molecular correlates of the specific vulnerability of the VLS, we analyzed the expression of voltage-gated potassium channels in rodent and primate brains using qPCR, in situ hybridization, and immunocytochemical single and double staining. Here we describe a novel, giant, non-cholinergic interneuron within the VLS. This neuron coexpresses the vesicular GABA transporter, the calcium-binding protein parvalbumin (PV), and the Kv3.3 potassium channel subunit. This novel neuron is much larger than PV neurons in other striatal regions, displays characteristic electrophysiological properties, and, most importantly, is restricted to the VLS. Consequently, the giant striatal Kv3.3-expressing PV neuron may link compromised Kv3 channel function and VLS-based orofacial dyskinesias.
Collapse
Affiliation(s)
- Lydia Lebenheim
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany
| | - Sam A Booker
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Christian Derst
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany
| | - Torsten Weiss
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany
| | - Franziska Wagner
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany.,Hans Berger Klinik für Neurologie, Universitätsklinikum Jena, An der Klinik 1, D-07747, Jena, Germany
| | - Clemens Gruber
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany
| | - Imre Vida
- Institut für Integrative Neuroanatomie, Charité-Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany
| | - Daniel S Zahm
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA.
| | - Rüdiger W Veh
- Institut für Zell- und Neurobiologie, Charité -Universitätsmedizin Berlin, Philippstraße 12, D-10115, Berlin, Germany.
| |
Collapse
|
11
|
Benarroch E. What Is the Role of Potassium Channels in Ataxia? Neurology 2021; 97:938-941. [PMID: 34782409 DOI: 10.1212/wnl.0000000000012870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 11/15/2022] Open
|
12
|
GAD67-mediated GABA Synthesis and Signaling Impinges on Directing Basket Cell Axonal Projections Toward Purkinje Cells in the Cerebellum. THE CEREBELLUM 2021; 21:905-919. [PMID: 34676525 DOI: 10.1007/s12311-021-01334-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
Gamma-aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the central nervous system, synthesized by two isoforms of glutamate decarboxylase (GAD): GAD65 and GAD67. GABA may act as a trophic factor during brain development, but its contribution to the development and maturation of cerebellar neural circuits is not known. To understand the roles of GABA in cerebellar organization and associated functions in motor coordination and balance, we examined GAD65 conventional knock out (KO) mice and mice in which GAD67 was eliminated in parvalbumin-expressing neurons (PV-Cre; GAD67flox/flox mice). We found aberrant subcellular localization of the Shaker-type K channel Kv1.1 in basket cell collaterals of PV-Cre; GAD67 flox/flox mice and abnormal projections from basket cells to Purkinje cells in both mouse strains. We also found that altered synaptic properties of basket cell terminals to Purkinje cells in PV-Cre; GAD67flox/flox mice. Furthermore, PV-Cre; GAD67 flox/flox mice exhibited abnormal motor coordination in the rotarod test. These results indicate that GABA signaling in the cerebellum is critical for establishing appropriate connections between basket cells and Purkinje cells and is associated with motor coordination in mice.
Collapse
|
13
|
Abstract
Transient outward potassium currents were first described nearly 60 years ago, since then major strides have been made in understanding their molecular basis and physiological roles. From the large family of voltage-gated potassium channels members of 3 subfamilies can produce such fast-inactivating A-type potassium currents. Each subfamily gives rise to currents with distinct biophysical properties and pharmacological profiles and a simple workflow is provided to aid the identification of channels mediating A-type currents in native cells. Their unique properties and regulation enable A-type K+ channels to perform varied roles in excitable cells including repolarisation of the cardiac action potential, controlling spike and synaptic timing, regulating dendritic integration and long-term potentiation as well as being a locus of neural plasticity.
Collapse
Affiliation(s)
- Jamie Johnston
- Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
14
|
Burger CA, Jiang D, Mackin RD, Samuel MA. Development and maintenance of vision's first synapse. Dev Biol 2021; 476:218-239. [PMID: 33848537 DOI: 10.1016/j.ydbio.2021.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/21/2022]
Abstract
Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.
Collapse
Affiliation(s)
- Courtney A Burger
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Robert D Mackin
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Bushart DD, Zalon AJ, Zhang H, Morrison LM, Guan Y, Paulson HL, Shakkottai VG, McLoughlin HS. Antisense Oligonucleotide Therapy Targeted Against ATXN3 Improves Potassium Channel-Mediated Purkinje Neuron Dysfunction in Spinocerebellar Ataxia Type 3. CEREBELLUM (LONDON, ENGLAND) 2021; 20:41-53. [PMID: 32789747 PMCID: PMC7930886 DOI: 10.1007/s12311-020-01179-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is the second-most common CAG repeat disease, caused by a glutamine-encoding expansion in the ATXN3 protein. SCA3 is characterized by spinocerebellar degeneration leading to progressive motor incoordination and early death. Previous studies suggest that potassium channel dysfunction underlies early abnormalities in cerebellar cortical Purkinje neuron firing in SCA3. However, cerebellar cortical degeneration is often modest both in the human disease and mouse models of SCA3, raising uncertainty about the role of cerebellar dysfunction in SCA3. Here, we address this question by investigating Purkinje neuron excitability in SCA3. In early-stage SCA3 mice, we confirm a previously identified increase in excitability of cerebellar Purkinje neurons and associate this excitability with reduced transcripts of two voltage-gated potassium (KV) channels, Kcna6 and Kcnc3, as well as motor impairment. Intracerebroventricular delivery of antisense oligonucleotides (ASO) to reduce mutant ATXN3 restores normal excitability to SCA3 Purkinje neurons and rescues transcript levels of Kcna6 and Kcnc3. Interestingly, while an even broader range of KV channel transcripts shows reduced levels in late-stage SCA3 mice, cerebellar Purkinje neuron physiology was not further altered despite continued worsening of motor impairment. These results suggest the progressive motor phenotype observed in SCA3 may not reflect ongoing changes in the cerebellar cortex but instead dysfunction of other neuronal structures within and beyond the cerebellum. Nevertheless, the early rescue of both KV channel expression and neuronal excitability by ASO treatment suggests that cerebellar cortical dysfunction contributes meaningfully to motor dysfunction in SCA3.
Collapse
Affiliation(s)
- David D. Bushart
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Annie J. Zalon
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Hongjiu Zhang
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109,Microsoft, Inc. Bellevue, WA 98004
| | - Logan M. Morrison
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109
| | - Yuanfang Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Henry L. Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Vikram G. Shakkottai
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109,Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109,Address correspondence to: Vikram G. Shakkottai, 4009 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, ; Hayley S. McLoughlin, 4017 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109,
| | - Hayley S. McLoughlin
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109,Address correspondence to: Vikram G. Shakkottai, 4009 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, ; Hayley S. McLoughlin, 4017 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109,
| |
Collapse
|
16
|
Samer S, Raman R, Laube G, Kreutz MR, Karpova A. The nuclear lamina is a hub for the nuclear function of Jacob. Mol Brain 2021; 14:9. [PMID: 33436037 PMCID: PMC7802242 DOI: 10.1186/s13041-020-00722-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Jacob is a synapto-nuclear messenger protein that couples NMDAR activity to CREB-dependent gene expression. In this study, we investigated the nuclear distribution of Jacob and report a prominent targeting to the nuclear envelope that requires NMDAR activity and nuclear import. Immunogold electron microscopy and proximity ligation assay combined with STED imaging revealed preferential association of Jacob with the inner nuclear membrane where it directly binds to LaminB1, an intermediate filament and core component of the inner nuclear membrane (INM). The association with the INM is transient; it involves a functional nuclear export signal in Jacob and a canonical CRM1-RanGTP-dependent export mechanism that defines the residing time of the protein at the INM. Taken together, the data suggest a stepwise redistribution of Jacob within the nucleus following nuclear import and prior to nuclear export.
Collapse
Affiliation(s)
- Sebastian Samer
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Rajeev Raman
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Gregor Laube
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, 39106, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| | - Anna Karpova
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, 39106, Magdeburg, Germany.
| |
Collapse
|
17
|
Koźmiński W, Pera J. Involvement of the Peripheral Nervous System in Episodic Ataxias. Biomedicines 2020; 8:biomedicines8110448. [PMID: 33105744 PMCID: PMC7690566 DOI: 10.3390/biomedicines8110448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/04/2022] Open
Abstract
Episodic ataxias comprise a group of inherited disorders, which have a common hallmark—transient attacks of ataxia. The genetic background is heterogeneous and the causative genes are not always identified. Furthermore, the clinical presentation, including intraictal and interictal symptoms, as well as the retention and progression of neurological deficits, is heterogeneous. Spells of ataxia can be accompanied by other symptoms—mostly from the central nervous system. However, in some of episodic ataxias involvement of peripheral nervous system is a part of typical clinical picture. This review intends to provide an insight into involvement of peripheral nervous system in episodic ataxias.
Collapse
Affiliation(s)
- Wojciech Koźmiński
- Department of Neurology, University Hospital, ul. Jakubowskiego 2, 30-688 Krakow, Poland;
| | - Joanna Pera
- Department of Neurology, Jagiellonian University Medical College, ul. Botaniczna 3, 31-503 Krakow, Poland
- Correspondence:
| |
Collapse
|
18
|
Zhou J, Brown AM, Lackey EP, Arancillo M, Lin T, Sillitoe RV. Purkinje cell neurotransmission patterns cerebellar basket cells into zonal modules defined by distinct pinceau sizes. eLife 2020; 9:55569. [PMID: 32990595 PMCID: PMC7561353 DOI: 10.7554/elife.55569] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 09/29/2020] [Indexed: 01/05/2023] Open
Abstract
Ramón y Cajal proclaimed the neuron doctrine based on circuit features he exemplified using cerebellar basket cell projections. Basket cells form dense inhibitory plexuses that wrap Purkinje cell somata and terminate as pinceaux at the initial segment of axons. Here, we demonstrate that HCN1, Kv1.1, PSD95 and GAD67 unexpectedly mark patterns of basket cell pinceaux that map onto Purkinje cell functional zones. Using cell-specific genetic tracing with an Ascl1CreERT2 mouse conditional allele, we reveal that basket cell zones comprise different sizes of pinceaux. We tested whether Purkinje cells instruct the assembly of inhibitory projections into zones, as they do for excitatory afferents. Genetically silencing Purkinje cell neurotransmission blocks the formation of sharp Purkinje cell zones and disrupts excitatory axon patterning. The distribution of pinceaux into size-specific zones is eliminated without Purkinje cell GABAergic output. Our data uncover the cellular and molecular diversity of a foundational synapse that revolutionized neuroscience.
Collapse
Affiliation(s)
- Joy Zhou
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, United States
| | - Amanda M Brown
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, United States
| | - Elizabeth P Lackey
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, United States
| | - Marife Arancillo
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, United States
| | - Tao Lin
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, United States
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| |
Collapse
|
19
|
Nam SM, Seo JS, Nahm SS, Chang BJ. Effects of ascorbic acid treatment on developmental alterations in calcium-binding proteins and gamma-aminobutyric acid transporter 1 in the cerebellum of lead-exposed rats during pregnancy and lactation. J Toxicol Sci 2020; 44:799-809. [PMID: 31708536 DOI: 10.2131/jts.44.799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In the present study, we investigated the effects of lead (Pb) and ascorbic acid co-administration on rat cerebellar development. Female rats were randomly divided into the following groups: control, Pb, and Pb plus ascorbic acid (PA) groups. From one week prior to mating, female rats were administered Pb (0.3% Pb acetate in drinking water) and ascorbic acid (100 mg/kg, oral intubation). The chemical administration was stopped on postnatal day 21 when the morphology of the offspring's cerebellum is similar to that of the adult brain. The blood Pb level was significantly increased following long-term Pb exposure. Ascorbic acid reduced Pb levels in the dams and offspring. Nissl staining demonstrated that the number of Purkinje cells was significantly reduced following Pb exposure, while ascorbic acid ameliorated this effect in the cerebellum of the offspring. Calcium-binding proteins, such as calbindin, calretinin, and parvalbumin were commonly expressed in Purkinje cells, and Pb exposure and ascorbic acid treatment resulted in similar patterns of change, namely Pb-induced impairment and ascorbic acid-mediated amelioration. The gamma-aminobutyric acid transporter 1 (GABAT1) is expressed in the pinceau structure where the somata of Purkinje cells are entwined in inhibitory synapses. The number of GABAT1-immunoreactive synapses was reduced following Pb exposure, and ascorbic acid co-treatment prevented this effect in the cerebellar cortex. Therefore, it can be concluded that ascorbic acid supplementation to mothers during gestation and lactation may have potential preventive effects against Pb-induced impairments in the developing cerebellum via protection of inhibitory neurons and synapses.
Collapse
Affiliation(s)
- Sung Min Nam
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Korea.,College of Veterinary Medicine and Veterinary Science Research Institute, Konkuk University, Korea
| | - Jin Seok Seo
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Korea
| | - Sang-Soep Nahm
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Korea.,College of Veterinary Medicine and Veterinary Science Research Institute, Konkuk University, Korea
| | - Byung-Joon Chang
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Korea.,College of Veterinary Medicine and Veterinary Science Research Institute, Konkuk University, Korea
| |
Collapse
|
20
|
Nozawa K, Hayashi A, Motohashi J, Takeo YH, Matsuda K, Yuzaki M. Cellular and Subcellular Localization of Endogenous Neuroligin-1 in the Cerebellum. THE CEREBELLUM 2018; 17:709-721. [PMID: 30046996 DOI: 10.1007/s12311-018-0966-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Synapses are precisely established, maintained, and modified throughout life by molecules called synaptic organizers, which include neurexins and neuroligins (Nlgn). Despite the importance of synaptic organizers in defining functions of neuronal circuits, the cellular and subcellular localization of many synaptic organizers has remained largely elusive because of the paucity of specific antibodies for immunohistochemical studies. In the present study, rather than raising specific antibodies, we generated knock-in mice in which a hemagglutinin (HA) epitope was inserted in the Nlgn1 gene. We have achieved high-throughput and precise gene editing by delivering the CRISPR/Cas9 system into zygotes. Using HA-Nlgn1 mice, we found that HA-Nlgn1 was enriched at synapses between parallel fibers and molecular layer interneurons (MLIs) and the glomeruli, in which mossy fiber terminals synapse onto granule cell dendrites. HA immunoreactivity was colocalized with postsynaptic density 95 at these synapses, indicating that endogenous Nlgn1 is localized at excitatory postsynaptic sites. In contrast, HA-Nlgn1 signals were very weak in dendrites and somata of Purkinje cells. Interestingly, HA-immunoreactivities were also observed in the pinceau, a specialized structure formed by MLI axons and astrocytes. HA-immunoreactivities in the pinceau were significantly reduced by knockdown of Nlgn1 in MLIs, indicating that in addition to postsynaptic sites, Nlgn1 is also localized at MLI axons. Our results indicate that epitope-tagging by electroporation-based gene editing with CRISPR/Cas9 is a viable and powerful method for mapping endogenous synaptic organizers with subcellular resolution, without the need for specific antibodies for each protein.
Collapse
Affiliation(s)
- Kazuya Nozawa
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Ayumi Hayashi
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Junko Motohashi
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yukari H Takeo
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Keiko Matsuda
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
21
|
Völker LA, Maar BA, Pulido Guevara BA, Bilkei-Gorzo A, Zimmer A, Brönneke H, Dafinger C, Bertsch S, Wagener JR, Schweizer H, Schermer B, Benzing T, Hoehne M. Neph2/Kirrel3 regulates sensory input, motor coordination, and home-cage activity in rodents. GENES BRAIN AND BEHAVIOR 2018; 17:e12516. [DOI: 10.1111/gbb.12516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/22/2018] [Accepted: 08/17/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Linus A. Völker
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Barbara A. Maar
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Barbara A. Pulido Guevara
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry; Medical Faculty of the University of Bonn; Bonn Germany
| | - Andreas Zimmer
- Institute of Molecular Psychiatry; Medical Faculty of the University of Bonn; Bonn Germany
| | - Hella Brönneke
- Mouse Phenotyping Core Facility; Cologne Excellence Cluster on Cellular Stress Responses (CECAD); 50931 Cologne Germany
| | - Claudia Dafinger
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Sabine Bertsch
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| | - Jan-Robin Wagener
- Institute for Neuroanatomy, Universitätsmedizin Göttingen; Georg-August-University Göttingen; Göttingen Germany
| | - Heiko Schweizer
- Renal Division; University Hospital Freiburg; Freiburg Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne Germany
- Systems Biology of Ageing Cologne (Sybacol); University of Cologne; Cologne Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne Germany
- Systems Biology of Ageing Cologne (Sybacol); University of Cologne; Cologne Germany
| | - Martin Hoehne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne; University of Cologne; Cologne Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne Germany
- Systems Biology of Ageing Cologne (Sybacol); University of Cologne; Cologne Germany
| |
Collapse
|
22
|
Hoxha E, Balbo I, Miniaci MC, Tempia F. Purkinje Cell Signaling Deficits in Animal Models of Ataxia. Front Synaptic Neurosci 2018; 10:6. [PMID: 29760657 PMCID: PMC5937225 DOI: 10.3389/fnsyn.2018.00006] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022] Open
Abstract
Purkinje cell (PC) dysfunction or degeneration is the most frequent finding in animal models with ataxic symptoms. Mutations affecting intrinsic membrane properties can lead to ataxia by altering the firing rate of PCs or their firing pattern. However, the relationship between specific firing alterations and motor symptoms is not yet clear, and in some cases PC dysfunction precedes the onset of ataxic signs. Moreover, a great variety of ionic and synaptic mechanisms can affect PC signaling, resulting in different features of motor dysfunction. Mutations affecting Na+ channels (NaV1.1, NaV1.6, NaVβ4, Fgf14 or Rer1) reduce the firing rate of PCs, mainly via an impairment of the Na+ resurgent current. Mutations that reduce Kv3 currents limit the firing rate frequency range. Mutations of Kv1 channels act mainly on inhibitory interneurons, generating excessive GABAergic signaling onto PCs, resulting in episodic ataxia. Kv4.3 mutations are responsible for a complex syndrome with several neurologic dysfunctions including ataxia. Mutations of either Cav or BK channels have similar consequences, consisting in a disruption of the firing pattern of PCs, with loss of precision, leading to ataxia. Another category of pathogenic mechanisms of ataxia regards alterations of synaptic signals arriving at the PC. At the parallel fiber (PF)-PC synapse, mutations of glutamate delta-2 (GluD2) or its ligand Crbl1 are responsible for the loss of synaptic contacts, abolishment of long-term depression (LTD) and motor deficits. At the same synapse, a correct function of metabotropic glutamate receptor 1 (mGlu1) receptors is necessary to avoid ataxia. Failure of climbing fiber (CF) maturation and establishment of PC mono-innervation occurs in a great number of mutant mice, including mGlu1 and its transduction pathway, GluD2, semaphorins and their receptors. All these models have in common the alteration of PC output signals, due to a variety of mechanisms affecting incoming synaptic signals or the way they are processed by the repertoire of ionic channels responsible for intrinsic membrane properties. Although the PC is a final common pathway of ataxia, the link between specific firing alterations and neurologic symptoms has not yet been systematically studied and the alterations of the cerebellar contribution to motor signals are still unknown.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Ilaria Balbo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy.,National Institute of Neuroscience (INN), Turin, Italy
| |
Collapse
|
23
|
Rowan MJM, Christie JM. Rapid State-Dependent Alteration in K v3 Channel Availability Drives Flexible Synaptic Signaling Dependent on Somatic Subthreshold Depolarization. Cell Rep 2017; 18:2018-2029. [PMID: 28228266 DOI: 10.1016/j.celrep.2017.01.068] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/16/2016] [Accepted: 01/25/2017] [Indexed: 10/20/2022] Open
Abstract
In many neurons, subthreshold depolarization in the soma can transiently increase action-potential (AP)-evoked neurotransmission via analog-to-digital facilitation. The mechanisms underlying this form of short-term synaptic plasticity are unclear, in part, due to the relative inaccessibility of the axon to direct physiological interrogation. Using voltage imaging and patch-clamp recording from presynaptic boutons of cerebellar stellate interneurons, we observed that depolarizing somatic potentials readily spread into the axon, resulting in AP broadening, increased spike-evoked Ca2+ entry, and enhanced neurotransmission strength. Kv3 channels, which drive AP repolarization, rapidly inactivated upon incorporation of Kv3.4 subunits. This leads to fast susceptibility to depolarization-induced spike broadening and analog facilitation independent of Ca2+-dependent protein kinase C signaling. The spread of depolarization into the axon was attenuated by hyperpolarization-activated currents (Ih currents) in the maturing cerebellum, precluding analog facilitation. These results suggest that analog-to-digital facilitation is tempered by development or experience in stellate cells.
Collapse
Affiliation(s)
- Matthew J M Rowan
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Jason M Christie
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA.
| |
Collapse
|
24
|
Fuchs JR, Darlington SW, Green JT, Morielli AD. Cerebellar learning modulates surface expression of a voltage-gated ion channel in cerebellar cortex. Neurobiol Learn Mem 2017; 142:252-262. [PMID: 28512010 DOI: 10.1016/j.nlm.2017.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/05/2017] [Accepted: 05/12/2017] [Indexed: 11/30/2022]
Abstract
Numerous experiments using ex vivo electrophysiology suggest that mammalian learning and memory involves regulation of voltage-gated ion channels in terms of changes in function. Yet, little is known about learning-related regulation of voltage-gated ion channels in terms of changes in expression. In two experiments, we examined changes in cell surface expression of the voltage-gated potassium channel alpha-subunit Kv1.2 in a discrete region of cerebellar cortex after eyeblink conditioning (EBC), a well-studied form of cerebellar-dependent learning. Kv1.2 in cerebellar cortex is expressed almost entirely in basket cells, primarily in the axon terminal pinceaux (PCX) region, and Purkinje cells, primarily in dendrites. Cell surface expression of Kv1.2 was measured using both multiphoton microscopy, which allowed measurement confined to the PCX region, and biotinylation/western blot, which measured total cell surface expression. In the first experiment, rats underwent three sessions of EBC, explicitly unpaired stimulus exposure, or context-only exposure and the results revealed a decrease in Kv1.2 cell surface expression in the unpaired group as measured with microscopy but no change as measured with western blot. In the second experiment, the same three training groups underwent only one half of a session of training, and the results revealed an increase in Kv1.2 cell surface expression in the unpaired group as measured with western blot but no change as measured with microscopy. In addition, rats in the EBC group that did not express conditioned responses (CRs) exhibited the same increase in Kv1.2 cell surface expression as the unpaired group. The overall pattern of results suggests that cell surface expression of Kv1.2 is changed with exposure to EBC stimuli in the absence, or prior to the emergence, of CRs.
Collapse
Affiliation(s)
- Jason R Fuchs
- Department of Psychological Science, University of Vermont, Burlington, VT 05405, United States
| | - Shelby W Darlington
- Department of Psychological Science, University of Vermont, Burlington, VT 05405, United States
| | - John T Green
- Department of Psychological Science, University of Vermont, Burlington, VT 05405, United States
| | - Anthony D Morielli
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, United States.
| |
Collapse
|
25
|
Harden N, Wang SJH, Krieger C. Making the connection – shared molecular machinery and evolutionary links underlie the formation and plasticity of occluding junctions and synapses. J Cell Sci 2016; 129:3067-76. [DOI: 10.1242/jcs.186627] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT
The pleated septate junction (pSJ), an ancient structure for cell–cell contact in invertebrate epithelia, has protein components that are found in three more-recent junctional structures, the neuronal synapse, the paranodal region of the myelinated axon and the vertebrate epithelial tight junction. These more-recent structures appear to have evolved through alterations of the ancestral septate junction. During its formation in the developing animal, the pSJ exhibits plasticity, although the final structure is extremely robust. Similar to the immature pSJ, the synapse and tight junctions both exhibit plasticity, and we consider evidence that this plasticity comes at least in part from the interaction of members of the immunoglobulin cell adhesion molecule superfamily with highly regulated membrane-associated guanylate kinases. This plasticity regulation probably arose in order to modulate the ancestral pSJ and is maintained in the derived structures; we suggest that it would be beneficial when studying plasticity of one of these structures to consider the literature on the others. Finally, looking beyond the junctions, we highlight parallels between epithelial and synaptic membranes, which both show a polarized distribution of many of the same proteins – evidence that determinants of apicobasal polarity in epithelia also participate in patterning of the synapse.
Collapse
Affiliation(s)
- Nicholas Harden
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, British Columbia V5A 1S6, Canada
| | - Simon Ji Hau Wang
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, British Columbia V5A 1S6, Canada
- Simon Fraser University, Department of Biomedical Physiology and Kinesiology, Burnaby, British Columbia V5A 1S6, Canada
| | - Charles Krieger
- Simon Fraser University, Department of Biomedical Physiology and Kinesiology, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
26
|
Begum R, Bakiri Y, Volynski KE, Kullmann DM. Action potential broadening in a presynaptic channelopathy. Nat Commun 2016; 7:12102. [PMID: 27381274 PMCID: PMC4935806 DOI: 10.1038/ncomms12102] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 05/27/2016] [Indexed: 12/31/2022] Open
Abstract
Brain development and interictal function are unaffected in many paroxysmal neurological channelopathies, possibly explained by homoeostatic plasticity of synaptic transmission. Episodic ataxia type 1 is caused by missense mutations of the potassium channel Kv1.1, which is abundantly expressed in the terminals of cerebellar basket cells. Presynaptic action potentials of small inhibitory terminals have not been characterized, and it is not known whether developmental plasticity compensates for the effects of Kv1.1 dysfunction. Here we use visually targeted patch-clamp recordings from basket cell terminals of mice harbouring an ataxia-associated mutation and their wild-type littermates. Presynaptic spikes are followed by a pronounced afterdepolarization, and are broadened by pharmacological blockade of Kv1.1 or by a dominant ataxia-associated mutation. Somatic recordings fail to detect such changes. Spike broadening leads to increased Ca2+ influx and GABA release, and decreased spontaneous Purkinje cell firing. We find no evidence for developmental compensation for inherited Kv1.1 dysfunction. Episodic ataxia type 1 is caused by mutations in the potassium channel Kv1.1, which is found in cerebellar basket cells. Here, the authors use electrophysiology techniques to characterize these mutant channels, and observe that the changes result in decreased spontaneous Purkinje cell firing with no evidence for developmental compensation.
Collapse
Affiliation(s)
- Rahima Begum
- UCL Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Yamina Bakiri
- UCL Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Kirill E Volynski
- UCL Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Dimitri M Kullmann
- UCL Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
27
|
Chihabi K, Morielli AD, Green JT. Intracerebellar infusion of the protein kinase M zeta (PKMζ) inhibitor zeta-inhibitory peptide (ZIP) disrupts eyeblink classical conditioning. Behav Neurosci 2016; 130:563-571. [PMID: 26949968 DOI: 10.1037/bne0000140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein kinase M zeta (PKM-ζ), a constitutively active N-terminal truncated form of PKC-ζ, has long been implicated in a cellular correlate of learning, long-term potentiation (LTP). Inhibition of PKM-ζ with zeta-inhibitory peptide (ZIP) has been shown in many brain structures to disrupt maintenance of AMPA receptors, irreversibly disrupting numerous forms of learning and memory that have been maintained for weeks. Delay eyeblink conditioning (EBC) is an established model for the assessment of cerebellar learning; here, we show that PKC-ζ and PKM-ζ are highly expressed in the cerebellar cortex, with highest expression found in Purkinje cell (PC) nuclei. Despite being highly expressed in the cerebellar cortex, no studies have examined how regulation of cerebellar PKM-ζ may affect cerebellar-dependent learning and memory. Given its disruption of learning in other brain structures, we hypothesized that ZIP would also disrupt delay EBC. We have shown that infusion of ZIP into the lobulus simplex of the rat cerebellar cortex can indeed significantly disrupt delay EBC. (PsycINFO Database Record
Collapse
|
28
|
Selective Loss of Presynaptic Potassium Channel Clusters at the Cerebellar Basket Cell Terminal Pinceau in Adam11 Mutants Reveals Their Role in Ephaptic Control of Purkinje Cell Firing. J Neurosci 2015; 35:11433-44. [PMID: 26269648 DOI: 10.1523/jneurosci.1346-15.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED A specialized axonal ending, the basket cell "pinceau," encapsulates the Purkinje cell axon initial segment (AIS), exerting final inhibitory control over the integrated outflow of the cerebellar cortex. This nonconventional axo-axonic contact extends beyond the perisomatic chemical GABAergic synaptic boutons to the distal AIS, lacks both sodium channels and local exocytotic machinery, and yet contains a dense cluster of voltage-gated potassium channels whose functional contribution is unknown. Here, we show that ADAM11, a transmembrane noncatalytic disintegrin, is the first reported Kv1-interacting protein essential for localizing Kv1.1 and Kv1.2 subunit complexes to the distal terminal. Selective absence of these channels at the pinceau due to mutation of ADAM11 spares spontaneous GABA release from basket cells at the perisomatic synapse yet eliminates ultrarapid ephaptic inhibitory synchronization of Purkinje cell firing. Our findings identify a critical role for presynaptic K(+) channels at the pinceau in ephaptic control over the speed and stability of spike rate coding at the Purkinje cell AIS in mice. SIGNIFICANCE STATEMENT This study identifies ADAM11 as the first essential molecule for the proper localization of potassium ion channels at presynaptic nerve terminals, where they modulate excitability and the release of neural transmitters. Genetic truncation of the transmembrane disintegrin and metalloproteinase protein ADAM11 resulted in the absence of Kv1 channels that are normally densely clustered at the terminals of basket cell axons in the cerebellar cortex. These specialized terminals are responsible for the release of the neurotransmitter GABA onto Purkinje cells and also display electrical signaling. In the ADAM11 mutant, GABAergic release was not altered, but the ultrarapid electrical signal was absent, demonstrating that the dense presynaptic cluster of Kv1 ion channels at these terminals mediate electrical transmission. Therefore, ADAM11 plays a critical role at this central synapse.
Collapse
|
29
|
Sotelo C. Molecular layer interneurons of the cerebellum: developmental and morphological aspects. CEREBELLUM (LONDON, ENGLAND) 2015; 14:534-56. [PMID: 25599913 DOI: 10.1007/s12311-015-0648-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During the past 25 years, our knowledge on the development of basket and stellate cells (molecular layer interneurons [MLIs]) has completely changed, not only regarding their origin from the ventricular zone, corresponding to the primitive cerebellar neuroepithelium, instead of the external granular layer, but above all by providing an almost complete account of the genetic regulations (transcription factors and other genes) involved in their differentiation and synaptogenesis. Moreover, it has been shown that MLIs' precursors (dividing neuroblasts) and not young postmitotic neurons, as in other germinal neuroepithelia, leave the germinative zone and migrate all along a complex and lengthy path throughout the presumptive cerebellar white matter, which provides suitable niches exerting epigenetic influences on their ultimate neuronal identities. Recent studies carried out on the anatomical-functional properties of adult MLIs emphasize the importance of these interneurons in regulating PC inhibition, and point out the crucial role played by electrical synaptic transmission between MLIs as well as ephaptic interactions between them and Purkinje cells at the pinceaux level, in the regulation of this inhibition.
Collapse
Affiliation(s)
- Constantino Sotelo
- INSERM, UMRS_U968, Institut de la Vision, 17 Rue Moreau, Paris, 75012, France.
- Institut de la Vision, Sorbonne Université, UPMC Univ Paris 06, Paris, 75012, France.
- CNRS, UMR_7210, Paris, 75012, France.
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Miguel Hernández (UMH), Avenida Ramón y Cajal s/n, 03550, San Juan de Alicante, Spain.
| |
Collapse
|
30
|
D'Adamo MC, Hasan S, Guglielmi L, Servettini I, Cenciarini M, Catacuzzeno L, Franciolini F. New insights into the pathogenesis and therapeutics of episodic ataxia type 1. Front Cell Neurosci 2015; 9:317. [PMID: 26347608 PMCID: PMC4541215 DOI: 10.3389/fncel.2015.00317] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/30/2015] [Indexed: 11/13/2022] Open
Abstract
Episodic ataxia type 1 (EA1) is a K+channelopathy characterized by a broad spectrum of symptoms. Generally, patients may experience constant myokymia and dramatic episodes of spastic contractions of the skeletal muscles of the head, arms, and legs with loss of both motor coordination and balance. During attacks additional symptoms may be reported such as vertigo, blurred vision, diplopia, nausea, headache, diaphoresis, clumsiness, stiffening of the body, dysarthric speech, and difficulty in breathing. These episodes may be precipitated by anxiety, emotional stress, fatigue, startle response or sudden postural changes. Epilepsy is overrepresented in EA1. The disease is inherited in an autosomal dominant manner, and genetic analysis of several families has led to the discovery of a number of point mutations in the voltage-dependent K+ channel gene KCNA1 (Kv1.1), on chromosome 12p13. To date KCNA1 is the only gene known to be associated with EA1. Functional studies have shown that these mutations impair Kv1.1 channel function with variable effects on channel assembly, trafficking and biophysics. Despite the solid evidence obtained on the molecular mechanisms underlying EA1, how these cause dysfunctions within the central and peripheral nervous systems circuitries remains elusive. This review summarizes the main breakthrough findings in EA1, discusses the neurophysiological mechanisms underlying the disease, current therapies, future challenges and opens a window onto the role of Kv1.1 channels in central nervous system (CNS) and peripheral nervous system (PNS) functions.
Collapse
Affiliation(s)
- Maria Cristina D'Adamo
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Sonia Hasan
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Luca Guglielmi
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Ilenio Servettini
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Marta Cenciarini
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| |
Collapse
|
31
|
Trimmer JS. Subcellular localization of K+ channels in mammalian brain neurons: remarkable precision in the midst of extraordinary complexity. Neuron 2015; 85:238-56. [PMID: 25611506 DOI: 10.1016/j.neuron.2014.12.042] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Potassium channels (KChs) are the most diverse ion channels, in part due to extensive combinatorial assembly of a large number of principal and auxiliary subunits into an assortment of KCh complexes. Their structural and functional diversity allows KChs to play diverse roles in neuronal function. Localization of KChs within specialized neuronal compartments defines their physiological role and also fundamentally impacts their activity, due to localized exposure to diverse cellular determinants of channel function. Recent studies in mammalian brain reveal an exquisite refinement of KCh subcellular localization. This includes axonal KChs at the initial segment, and near/within nodes of Ranvier and presynaptic terminals, dendritic KChs found at sites reflecting specific synaptic input, and KChs defining novel neuronal compartments. Painting the remarkable diversity of KChs onto the complex architecture of mammalian neurons creates an elegant picture of electrical signal processing underlying the sophisticated function of individual neuronal compartments, and ultimately neurotransmission and behavior.
Collapse
Affiliation(s)
- James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
32
|
D’Adamo MC, Di Giovanni G, Pessia M. Animal Models of Episodic Ataxia Type 1 (EA1). Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00051-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
33
|
Fuchs JR, Robinson GM, Dean AM, Schoenberg HE, Williams MR, Morielli AD, Green JT. Cerebellar secretin modulates eyeblink classical conditioning. ACTA ACUST UNITED AC 2014; 21:668-75. [PMID: 25403455 PMCID: PMC4236411 DOI: 10.1101/lm.035766.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have previously shown that intracerebellar infusion of the neuropeptide secretin enhances the acquisition phase of eyeblink conditioning (EBC). Here, we sought to test whether endogenous secretin also regulates EBC and to test whether the effect of exogenous and endogenous secretin is specific to acquisition. In Experiment 1, rats received intracerebellar infusions of the secretin receptor antagonist 5-27 secretin or vehicle into the lobulus simplex of cerebellar cortex immediately prior to sessions 1-3 of acquisition. Antagonist-infused rats showed a reduction in the percentage of eyeblink CRs compared with vehicle-infused rats. In Experiment 2, rats received intracerebellar infusions of secretin or vehicle immediately prior to sessions 1-2 of extinction. Secretin did not significantly affect extinction performance. In Experiment 3, rats received intracerebellar infusions of 5-27 secretin or vehicle immediately prior to sessions 1-2 of extinction. The secretin antagonist did not significantly affect extinction performance. Together, our current and previous results indicate that both exogenous and endogenous cerebellar secretin modulate acquisition, but not extinction, of EBC. We have previously shown that (1) secretin reduces surface expression of the voltage-gated potassium channel α-subunit Kv1.2 in cerebellar cortex and (2) intracerebellar infusions of a Kv1.2 blocker enhance EBC acquisition, much like secretin. Kv1.2 is almost exclusively expressed in cerebellar cortex at basket cell-Purkinje cell pinceaus and Purkinje cell dendrites; we propose that EBC-induced secretin release from PCs modulates EBC acquisition by reducing surface expression of Kv1.2 at one or both of these sites.
Collapse
Affiliation(s)
- Jason R Fuchs
- Department of Psychology, University of Vermont, Burlington, Vermont 05405, USA
| | - Gain M Robinson
- Department of Psychology, University of Vermont, Burlington, Vermont 05405, USA
| | - Aaron M Dean
- Department of Psychology, University of Vermont, Burlington, Vermont 05405, USA
| | - Heidi E Schoenberg
- Department of Psychology, University of Vermont, Burlington, Vermont 05405, USA
| | - Michael R Williams
- Department of Pharmacology, University of Vermont, Burlington, Vermont 05405, USA
| | - Anthony D Morielli
- Department of Pharmacology, University of Vermont, Burlington, Vermont 05405, USA
| | - John T Green
- Department of Psychology, University of Vermont, Burlington, Vermont 05405, USA
| |
Collapse
|
34
|
Distinct Kv channel subtypes contribute to differences in spike signaling properties in the axon initial segment and presynaptic boutons of cerebellar interneurons. J Neurosci 2014; 34:6611-23. [PMID: 24806686 DOI: 10.1523/jneurosci.4208-13.2014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The discrete arrangement of voltage-gated K(+) (Kv) channels in axons may impart functional advantages in action potential (AP) signaling yet, in compact cell types, the organization of Kv channels is poorly understood. We find that in cerebellar stellate cell interneurons of mice, the composition and influence of Kv channels populating the axon is diverse and depends on location allowing axonal compartments to differentially control APs in a local manner. Kv1 channels determine AP repolarization at the spike initiation site but not at more distal sites, limiting the expression of use-dependent spike broadening to the most proximal axon region, likely a key attribute informing spiking phenotype. Local control of AP repolarization at presynaptic boutons depends on Kv3 channels keeping APs brief, thus limiting Ca(2+) influx and synaptic strength. These observations suggest that AP repolarization is tuned by the local influence of distinct Kv channel types, and this organization enhances the functional segregation of axonal compartments.
Collapse
|
35
|
Kim KY, Scholl ES, Liu X, Shepherd A, Haeseleer F, Lee A. Localization and expression of CaBP1/caldendrin in the mouse brain. Neuroscience 2014; 268:33-47. [PMID: 24631676 DOI: 10.1016/j.neuroscience.2014.02.052] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 12/31/2022]
Abstract
Ca(2+) binding protein 1 (CaBP1) and caldendrin are alternatively spliced variants of a subfamily of CaBPs with high homology to calmodulin. Although CaBP1 and caldendrin regulate effectors including plasma membrane and intracellular Ca(2+) channels in heterologous expression systems, little is known about their functions in vivo. Therefore, we generated mice deficient in CaBP1/caldendrin expression (C-KO) and analyzed the expression and cellular localization of CaBP1 and caldendrin in the mouse brain. Immunoperoxidase labeling with antibodies recognizing both CaBP1 and caldendrin was absent in the brain of C-KO mice, but was intense in multiple brain regions of wild-type mice. By Western blot, the antibodies detected two proteins that were absent in the C-KO mouse and consistent in size with caldendrin variants originating from alternative translation initiation sites. By quantitative PCR, caldendrin transcript levels were far greater than those for CaBP1, particularly in the cerebral cortex and hippocampus. In the frontal cortex but not in the hippocampus, caldendrin expression increased steadily from birth. By double-label immunofluorescence, CaBP1/caldendrin was localized in principal neurons and parvalbumin-positive interneurons. In the cerebellum, CaBP1/caldendrin antibodies labeled interneurons in the molecular layer and in basket cell terminals surrounding the soma and axon initial segment of Purkinje neurons, but immunolabeling was absent in Purkinje neurons. We conclude that CaBP1/caldendrin is localized both pre- and postsynaptically where it may regulate Ca(2+) signaling and excitability in select groups of excitatory and inhibitory neurons.
Collapse
Affiliation(s)
- K Y Kim
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - E S Scholl
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - X Liu
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - A Shepherd
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - F Haeseleer
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - A Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
36
|
Blot A, Barbour B. Ultra-rapid axon-axon ephaptic inhibition of cerebellar Purkinje cells by the pinceau. Nat Neurosci 2014; 17:289-95. [PMID: 24413696 DOI: 10.1038/nn.3624] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/10/2013] [Indexed: 11/09/2022]
Abstract
Excitatory synaptic activity in the brain is shaped and balanced by inhibition. Because inhibition cannot propagate, it is often recruited with a synaptic delay by incoming excitation. Cerebellar Purkinje cells are driven by long-range excitatory parallel fiber inputs, which also recruit local inhibitory basket cells. The axon initial segment of each Purkinje cell is ensheathed by basket cell axons in a structure called the pinceau, which is largely devoid of chemical synapses. In mice, we found at the single-cell level that the pinceau mediates ephaptic inhibition of Purkinje cell firing at the site of spike initiation. The reduction of firing rate was synchronous with the presynaptic action potential, eliminating a synaptic delay and allowing granule cells to inhibit Purkinje cells without a preceding phase of excitation. Axon-axon ephaptic intercellular signaling can therefore mediate near-instantaneous feedforward and lateral inhibition.
Collapse
Affiliation(s)
- Antonin Blot
- 1] Ecole Normale Supérieure, IBENS, Paris, France. [2] CNRS, UMR 8197, Paris, France. [3] INSERM, U1024, Paris, France
| | - Boris Barbour
- 1] Ecole Normale Supérieure, IBENS, Paris, France. [2] CNRS, UMR 8197, Paris, France. [3] INSERM, U1024, Paris, France
| |
Collapse
|
37
|
Martínez-Hernández J, Ballesteros-Merino C, Fernández-Alacid L, Nicolau JC, Aguado C, Luján R. Polarised localisation of the voltage-gated sodium channel Na(v)1.2 in cerebellar granule cells. THE CEREBELLUM 2013; 12:16-26. [PMID: 22528969 DOI: 10.1007/s12311-012-0387-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium channels are responsible for action potential initiation and propagation in electrically excitable cells. In this study, we used biochemical, immunohistochemical and quantitative immunoelectron microscopy techniques to reveal the temporal and spatial expression of the Na(v)1.2 channel subunit in granule cells of cerebellum. Using histoblot, we detected Na(v)1.2 widely distributed in the adult brain, but prominently expressed in the cerebellum. During postnatal development, Na(v)1.2 mRNA and protein were detected low during the first and second postnatal week, increased to P15 and then continue to decrease until adult levels. At the light microscopic level, Na(v)1.2 immunoreactivity concentrated in the molecular layer of the cerebellar cortex. Using immunofluorescence, Na(v)1.2 colocalised with VGluT1, but not with VGluT2, demonstrating that the subunit was preferentially present in parallel fibre axons and axon terminals. At the electron microscopic level, Na(v)1.2 immunoparticles were exclusively detected at presynaptic sites in granule cell axons and axon terminals of granule cells, with occasional clustering in their axon initial segment. This was demonstrated using quantitative immunogold analysis. In the axon terminals, the distribution of Na(v)1.2 was relatively uniform along the extrasynaptic plasma membrane and never detected in the active zone. We could not find detectable levels of Na(v)1.2 at postsynaptic elements of granule cells or other cerebellar cell types. The present findings show a polarised distribution of Na(v)1.2 along the neuronal surface of granule cells and suggest its primary involvement in the transmission of information from granule cells to Purkinje cells.
Collapse
Affiliation(s)
- José Martínez-Hernández
- Department of Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02006, Albacete, Spain
| | | | | | | | | | | |
Collapse
|
38
|
Buttermore ED, Thaxton CL, Bhat MA. Organization and maintenance of molecular domains in myelinated axons. J Neurosci Res 2013; 91:603-22. [PMID: 23404451 DOI: 10.1002/jnr.23197] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 09/19/2012] [Accepted: 11/28/2012] [Indexed: 01/17/2023]
Abstract
Over a century ago, Ramon y Cajal first proposed the idea of a directionality involved in nerve conduction and neuronal communication. Decades later, it was discovered that myelin, produced by glial cells, insulated axons with periodic breaks where nodes of Ranvier (nodes) form to allow for saltatory conduction. In the peripheral nervous system (PNS), Schwann cells are the glia that can either individually myelinate the axon from one neuron or ensheath axons of many neurons. In the central nervous system (CNS), oligodendrocytes are the glia that myelinate axons from different neurons. Review of more recent studies revealed that this myelination created polarized domains adjacent to the nodes. However, the molecular mechanisms responsible for the organization of axonal domains are only now beginning to be elucidated. The molecular domains in myelinated axons include the axon initial segment (AIS), where various ion channels are clustered and action potentials are initiated; the node, where sodium channels are clustered and action potentials are propagated; the paranode, where myelin loops contact with the axolemma; the juxtaparanode (JXP), where delayed-rectifier potassium channels are clustered; and the internode, where myelin is compactly wrapped. Each domain contains a unique subset of proteins critical for the domain's function. However, the roles of these proteins in axonal domain organization are not fully understood. In this review, we highlight recent advances on the molecular nature and functions of some of the components of each axonal domain and their roles in axonal domain organization and maintenance for proper neuronal communication.
Collapse
Affiliation(s)
- Elizabeth D Buttermore
- Curriculum in Neurobiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
39
|
Batsios P, Baumann O, Gräf R, Meyer I. Isolation of Dictyostelium nuclei for light and electron microscopy. Methods Mol Biol 2013; 983:283-294. [PMID: 23494313 DOI: 10.1007/978-1-62703-302-2_15] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The significance of amoebae for studies of nuclear architecture has considerably increased in the recent years. The availability of a protocol for isolation of nuclei in a quality sufficient for high-resolution light and electron microscopy is a prerequisite for such studies. Here we present a protocol for high enrichment of nuclei by sucrose density-gradient centrifugation. Moreover, we describe how to use these isolated nuclei as specimens for immunofluorescence and immune-electron microscopy of ultrathin sections.
Collapse
Affiliation(s)
- Petros Batsios
- Department of Cell Biology, Institute for Biochemistry and Biology, University of Potsdam, Potsdam-Golm, Germany
| | | | | | | |
Collapse
|
40
|
Cellular mechanisms and behavioral consequences of Kv1.2 regulation in the rat cerebellum. J Neurosci 2012; 32:9228-37. [PMID: 22764231 DOI: 10.1523/jneurosci.6504-11.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The potassium channel Kv1.2 α-subunit is expressed in cerebellar Purkinje cell (PC) dendrites where its pharmacological inhibition increases excitability (Khavandgar et al., 2005). Kv1.2 is also expressed in cerebellar basket cell (BC) axon terminals (Sheng et al., 1994), where its blockade increases BC inhibition of PCs (Southan and Robertson, 1998a). Secretin receptors are also expressed both in PC dendrites and BC axon terminals (for review, see (Yuan et al., 2011). The effect of secretin on PC excitability is not yet known, but, like Kv1.2 inhibitors, secretin potently increases inhibitory input to PCs (Yung et al., 2001). This suggests secretin may act in part by suppressing Kv1.2. Receptor-mediated endocytosis is a mechanism of Kv1.2 suppression (Nesti et al., 2004). This process can be regulated by protein kinase A (PKA) (Connors et al., 2008). Since secretin receptors activate PKA (Wessels-Reiker et al., 1993), we tested the hypothesis that secretin regulates Kv1.2 trafficking in the cerebellum. Using cell-surface protein biotinylation of rat cerebellar slices, we found secretin decreased cell-surface Kv1.2 levels by modulating Kv1.2 endocytic trafficking. This effect was mimicked by activating adenylate cyclase (AC) with forskolin, and was blocked by pharmacological inhibitors of AC or PKA. Imaging studies identified the BC axon terminal and PC dendrites as loci of AC-dependent Kv1.2 trafficking. The physiological significance of secretin-regulated Kv1.2 endocytosis is supported by our finding that infusion into the cerebellar cortex of either the Kv1.2 inhibitor tityustoxin-Kα, or of the Kv1.2 regulator secretin, significantly enhances acquisition of eyeblink conditioning in rats.
Collapse
|
41
|
Lack of molecular-anatomical evidence for GABAergic influence on axon initial segment of cerebellar Purkinje cells by the pinceau formation. J Neurosci 2012; 32:9438-48. [PMID: 22764252 DOI: 10.1523/jneurosci.1651-12.2012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The axon initial segment (AIS) of cerebellar Purkinje cells (PCs) is embraced by ramified axons of GABAergic basket cells (BCs) called the pinceau formation. This unique structure has been assumed to be a device for the modulation of PC outputs through electrical and/or GABAergic inhibition. Electrical inhibition is supported by enriched potassium channels, absence of sodium channels, and developed septate-like junctions between BC axons. The neurochemical basis for GABAergic inhibition, however, has not been well investigated. Here we addressed this issue using C56BL/6 mice. First, we confirmed previous observations that typical synaptic contacts were rare and confined to proximal axonal portions, with the remaining portions being mostly covered by astrocytic processes. Then we examined the expression of molecules involved in GABAergic signaling, including GABA synthetic enzyme glutamic acid decarboxylase (GAD), vesicular GABA transporter vesicular inhibitory amino acid transporter (VIAAT), cytomatrix active zone protein bassoon, GABA receptor GABA(A)Rα1, and cell adhesion molecule neuroligin-2. These molecules were recruited to form a functional assembly at perisomatic BC-PC synapses and along the AIS of hippocampal and neocortical pyramidal cells. GAD and VIAAT immunogold labeling was five times lower in the pinceau formation compared with perisomatic BC terminals and showed no accumulation toward the AIS. Moreover, bassoon, neuroligin-2, and GABA(A)Rα1 formed no detectable clusters along the ankyrin-G-positive AIS proper. These findings indicate that GABAergic signaling machinery is organized loosely and even incompletely in the pinceau formation. Together, BCs do not appear to exert GABAergic synaptic inhibition on the AIS, although the mode of action of the pinceau formation remains to be explored.
Collapse
|
42
|
Huang CY, Chu D, Hwang WC, Tsaur ML. Coexpression of high-voltage-activated ion channels Kv3.4 and Cav1.2 in pioneer axons during pathfinding in the developing rat forebrain. J Comp Neurol 2012; 520:3650-72. [DOI: 10.1002/cne.23119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Leitinger G, Masich S, Neumüller J, Pabst MA, Pavelka M, Rind FC, Shupliakov O, Simmons PJ, Kolb D. Structural organization of the presynaptic density at identified synapses in the locust central nervous system. J Comp Neurol 2012; 520:384-400. [PMID: 21826661 PMCID: PMC3263340 DOI: 10.1002/cne.22744] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In a synaptic active zone, vesicles aggregate around a densely staining structure called the presynaptic density. We focus on its three-dimensional architecture and a major molecular component in the locust. We used electron tomography to study the presynaptic density in synapses made in the brain by identified second-order neuron of the ocelli. Here, vesicles close to the active zone are organized in two rows on either side of the presynaptic density, a level of organization not previously reported in insect central synapses. The row of vesicles that is closest to the density's base includes vesicles docked with the presynaptic membrane and thus presumably ready for release, whereas the outer row of vesicles does not include any that are docked. We show that a locust ortholog of the Drosophila protein Bruchpilot is localized to the presynaptic density, both in the ocellar pathway and compound eye visual neurons. An antibody recognizing the C-terminus of the Bruchpilot ortholog selectively labels filamentous extensions of the presynaptic density that reach out toward vesicles. Previous studies on Bruchpilot have focused on its role in neuromuscular junctions in Drosophila, and our study shows it is also a major functional component of presynaptic densities in the central nervous system of an evolutionarily distant insect. Our study thus reveals Bruchpilot executes similar functions in synapses that can sustain transmission of small graded potentials as well as those relaying large, spike-evoked signals.
Collapse
Affiliation(s)
- Gerd Leitinger
- Institute of Cell Biology, Histology and Embryology, Center for Molecular Medicine (ZMM), Medical University of Graz, Austria. Gerd.Leitinger@medunigraz
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bucher D, Goaillard JM. Beyond faithful conduction: short-term dynamics, neuromodulation, and long-term regulation of spike propagation in the axon. Prog Neurobiol 2011; 94:307-46. [PMID: 21708220 PMCID: PMC3156869 DOI: 10.1016/j.pneurobio.2011.06.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 05/27/2011] [Accepted: 06/07/2011] [Indexed: 12/13/2022]
Abstract
Most spiking neurons are divided into functional compartments: a dendritic input region, a soma, a site of action potential initiation, an axon trunk and its collaterals for propagation of action potentials, and distal arborizations and terminals carrying the output synapses. The axon trunk and lower order branches are probably the most neglected and are often assumed to do nothing more than faithfully conducting action potentials. Nevertheless, there are numerous reports of complex membrane properties in non-synaptic axonal regions, owing to the presence of a multitude of different ion channels. Many different types of sodium and potassium channels have been described in axons, as well as calcium transients and hyperpolarization-activated inward currents. The complex time- and voltage-dependence resulting from the properties of ion channels can lead to activity-dependent changes in spike shape and resting potential, affecting the temporal fidelity of spike conduction. Neural coding can be altered by activity-dependent changes in conduction velocity, spike failures, and ectopic spike initiation. This is true under normal physiological conditions, and relevant for a number of neuropathies that lead to abnormal excitability. In addition, a growing number of studies show that the axon trunk can express receptors to glutamate, GABA, acetylcholine or biogenic amines, changing the relative contribution of some channels to axonal excitability and therefore rendering the contribution of this compartment to neural coding conditional on the presence of neuromodulators. Long-term regulatory processes, both during development and in the context of activity-dependent plasticity may also affect axonal properties to an underappreciated extent.
Collapse
Affiliation(s)
- Dirk Bucher
- The Whitney Laboratory and Department of Neuroscience, University of Florida, St. Augustine, FL 32080, USA.
| | | |
Collapse
|
45
|
Abstract
Axons are generally considered as reliable transmission cables in which stable propagation occurs once an action potential is generated. Axon dysfunction occupies a central position in many inherited and acquired neurological disorders that affect both peripheral and central neurons. Recent findings suggest that the functional and computational repertoire of the axon is much richer than traditionally thought. Beyond classical axonal propagation, intrinsic voltage-gated ionic currents together with the geometrical properties of the axon determine several complex operations that not only control signal processing in brain circuits but also neuronal timing and synaptic efficacy. Recent evidence for the implication of these forms of axonal computation in the short-term dynamics of neuronal communication is discussed. Finally, we review how neuronal activity regulates both axon morphology and axonal function on a long-term time scale during development and adulthood.
Collapse
Affiliation(s)
- Dominique Debanne
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Emilie Campanac
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Andrzej Bialowas
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Edmond Carlier
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Gisèle Alcaraz
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| |
Collapse
|
46
|
Luján R. Organisation of potassium channels on the neuronal surface. J Chem Neuroanat 2010; 40:1-20. [PMID: 20338235 DOI: 10.1016/j.jchemneu.2010.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 03/10/2010] [Accepted: 03/11/2010] [Indexed: 11/30/2022]
Abstract
Potassium channels are a family of ion channels that govern the intrinsic electrical properties of neurons in the brain. Molecular cloning has revealed over 100 genes encoding the pore-forming alpha subunits of potassium channels in mammals, making them the most diverse subset of ion channels. Multiplicity in this ion channel family is further generated through alternative splicing. The precise location of potassium channels along the dendro-somato-axonic surface of the neurons is an important factor in determining its functional impact. Today, it is widely accepted that potassium channels can be located at any subcellular compartment on the neuronal surface, at synaptic and extrasynaptic sites, from somata to dendritic shafts, dendritic spines, axons or axon terminals. However, they are not evenly distributed on the neuronal surface and depending on the potassium channel subtype, are instead concentrated at different compartments. This selective localization of ion channels to specific neuronal compartments has many different functional implications. One factor necessary to understand the role of potassium channels in neuronal function is to unravel their specialized distribution and subcellular localization within a cell, and this can only be achieved by electron microscopy. In this review, I summarize anatomical findings, describing their distribution in the central nervous system. The distinct regional, cellular and subcellular distribution of potassium channels in the brain will be discussed in view of their possible functional implications.
Collapse
Affiliation(s)
- Rafael Luján
- Departamento de Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02006 Albacete, Spain.
| |
Collapse
|
47
|
ADAM22, a Kv1 channel-interacting protein, recruits membrane-associated guanylate kinases to juxtaparanodes of myelinated axons. J Neurosci 2010; 30:1038-48. [PMID: 20089912 DOI: 10.1523/jneurosci.4661-09.2010] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Clustered Kv1 K(+) channels regulate neuronal excitability at juxtaparanodes of myelinated axons, axon initial segments, and cerebellar basket cell terminals (BCTs). These channels are part of a larger protein complex that includes cell adhesion molecules and scaffolding proteins. To identify proteins that regulate assembly, clustering, and/or maintenance of axonal Kv1 channel protein complexes, we immunoprecipitated Kv1.2 alpha subunits, and then used mass spectrometry to identify interacting proteins. We found that a disintegrin and metalloproteinase 22 (ADAM22) is a component of the Kv1 channel complex and that ADAM22 coimmunoprecipitates Kv1.2 and the membrane-associated guanylate kinases (MAGUKs) PSD-93 and PSD-95. When coexpressed with MAGUKs in heterologous cells, ADAM22 and Kv1 channels are recruited into membrane surface clusters. However, coexpression of Kv1.2 with ADAM22 and MAGUKs does not alter channel properties. Among all the known Kv1 channel-interacting proteins, only ADAM22 is found at every site where Kv1 channels are clustered. Analysis of Caspr-null mice showed that, like other previously described juxtaparanodal proteins, disruption of the paranodal junction resulted in redistribution of ADAM22 into paranodal zones. Analysis of Caspr2-, PSD-93-, PSD-95-, and double PSD-93/PSD-95-null mice showed ADAM22 clustering at BCTs requires PSD-95, but ADAM22 clustering at juxtaparanodes requires neither PSD-93 nor PSD-95. In direct contrast, analysis of ADAM22-null mice demonstrated juxtaparanodal clustering of PSD-93 and PSD-95 requires ADAM22, whereas Kv1.2 and Caspr2 clustering is normal in ADAM22-null mice. Thus, ADAM22 is an axonal component of the Kv1 K(+) channel complex that recruits MAGUKs to juxtaparanodes.
Collapse
|
48
|
Schulz I, Baumann O, Samereier M, Zoglmeier C, Gräf R. Dictyostelium Sun1 is a dynamic membrane protein of both nuclear membranes and required for centrosomal association with clustered centromeres. Eur J Cell Biol 2009; 88:621-38. [PMID: 19632001 DOI: 10.1016/j.ejcb.2009.06.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/23/2009] [Accepted: 06/26/2009] [Indexed: 11/25/2022] Open
Abstract
Centrosomal attachment to nuclei is crucial for proper mitosis and nuclear positioning in various organisms, and generally involves Sun-family proteins located at the inner nuclear envelope. There is still no common scheme for the outer nuclear membrane proteins interacting with Sun1 in centrosome/nucleus attachment. Here we propose a model in which Sun1 mediates a physical link between centrosomes and clustered centromeres through both nuclear membranes in Dictyostelium. For the first time we provide a detailed microscopic analysis of the centrosomal and nuclear envelope localization of endogenous Dictyostelium Sun1 during interphase and mitosis. By immunogold electron microscopy we show that Sun1 is a resident of both nuclear membranes. Disruption of Sun1 function by overexpression of full-length GFP-Sun1 or a GFP-Sun-domain deletion construct revealed not only the established function in centrosome/nucleus attachment and maintenance of ploidy, but also a requirement of Sun1 for the association of the centromere cluster with the centrosome. Live-cell imaging visualized the occurrence of mitotic defects, and demonstrated the requirement of microtubules for dynamic distance changes between centrosomes and nuclei. FRAP analysis revealed at least two populations of Sun1, with an immobile fraction associated with the centrosome, and a mobile fraction in the nuclear envelope.
Collapse
Affiliation(s)
- Irene Schulz
- Department of Cell Biology, Institute for Biochemistry and Biology, University of Potsdam, Potsdam-Golm, Germany
| | | | | | | | | |
Collapse
|
49
|
Stirling L, Williams MR, Morielli AD. Dual roles for RHOA/RHO-kinase in the regulated trafficking of a voltage-sensitive potassium channel. Mol Biol Cell 2009; 20:2991-3002. [PMID: 19403695 DOI: 10.1091/mbc.e08-10-1074] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Kv1.2 is a member of the Shaker family of voltage-sensitive potassium channels and contributes to regulation of membrane excitability. The electrophysiological activity of Kv1.2 undergoes tyrosine kinase-dependent suppression in a process involving RhoA. We report that RhoA elicits suppression of Kv1.2 ionic current by modulating channel endocytosis. This occurs through two distinct pathways, one clathrin-dependent and the other cholesterol-dependent. Activation of Rho kinase (ROCK) via the lysophosphatidic acid (LPA) receptor elicits clathrin-dependent Kv1.2 endocytosis and consequent attenuation of its ionic current. LPA-induced channel endocytosis is blocked by the ROCK inhibitor Y27632 or by clathrin RNA interference. In contrast, steady-state endocytosis of Kv1.2 in unstimulated cells is cholesterol dependent. Inhibition of basal ROCK signaling with Y27632 increased surface Kv1.2, an effect that persists in the presence of clathrin small interfering RNA and that is not additive to the increase in surface channel levels elicited by the cholesterol sequestering drug filipin. Temperature block experiments show that ROCK affects cholesterol-dependent trafficking by modulating the recycling of endocytosed channel back to the plasma membrane. Both receptor-stimulated and steady-state Kv1.2 trafficking modulated by RhoA/ROCK required the activation of dynamin as well as the ROCK effector Lim-kinase, indicating a key role for actin remodeling in RhoA-dependent Kv1.2 regulation.
Collapse
Affiliation(s)
- Lee Stirling
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | |
Collapse
|
50
|
Joho RH, Hurlock EC. The role of Kv3-type potassium channels in cerebellar physiology and behavior. THE CEREBELLUM 2009; 8:323-33. [PMID: 19247732 DOI: 10.1007/s12311-009-0098-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 02/10/2009] [Indexed: 10/21/2022]
Abstract
Different subunits of the Kv3 subfamily of voltage-gated potassium (Kv) channels (Kv3.1-Kv3.4) are expressed in distinct neuronal subpopulations in the cerebellum. Behavioral phenotypes in Kv3-null mutant mice such as ataxia with prominent hypermetria and heightened alcohol sensitivity are characteristic of cerebellar dysfunction. Here, we review how the unique biophysical properties of Kv3-type potassium channels, fast activation and fast deactivation that enable cerebellar neurons to generate brief action potentials at high frequencies, affect firing patterns and influence cerebellum-mediated behavior.
Collapse
Affiliation(s)
- Rolf H Joho
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | | |
Collapse
|