1
|
Isaac Guillén F, Geist MA, Cheng SY, Harris AM, Treviño ME, Brumback AC, Nishiyama H, Howard MA. A novel mouse model for developmental and epileptic encephalopathy by Purkinje cell-specific deletion of Scn1b. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624370. [PMID: 39605540 PMCID: PMC11601654 DOI: 10.1101/2024.11.19.624370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Loss of function variants of SCN1B are associated with a range of developmental and epileptic encephalopathies (DEEs), including Dravet syndrome. These DEEs feature a wide range of severe neurological disabilities, including changes to social, motor, mood, sleep, and cognitive function which are notoriously difficult to treat, and high rates of early mortality. While the symptomology of SCN1B -associated DEEs indicates broad changes in neural function, most research has focused on epilepsy-related brain structures and function. Mechanistic studies of SCN1B / Scn1b have delineated diverse roles in development and adult maintenance of neural function, via cell adhesion, ion channel regulation, and other intra- and extra-cellular actions. However, use of mouse models is limited as knockout of Scn1b , globally and even in some cell-specific models (e.g., Parvalbumin+ interneuron-specific knockout) in adult mice, leads to severe and progressive epilepsy, health deterioration, and 100% mortality within weeks. Here, we report findings of a novel transgenic mouse line in which Scn1b was specifically deleted in cerebellar Purkinje cells. Unlike most existing models, these mice did not show failure to thrive or early mortality. However, we quantified marked decrements to Purkinje cell physiology as well as motor, social, and cognitive dysfunction. Our data indicate that cerebellar Purkinje cells are an important node for dysfunction and neural disabilities in SCN1B -related DEEs, and combined with previous work identify this as a potentially vital site for understanding mechanisms of DEEs and developing therapies that can treat these disorders holistically.
Collapse
|
2
|
Bean BP. Mechanisms of pacemaking in mammalian neurons. J Physiol 2024. [PMID: 39303139 DOI: 10.1113/jp284759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Many neurons in the mammalian brain show pacemaking activity: rhythmic generation of action potentials in the absence of sensory or synaptic input. Slow pacemaking of neurons releasing modulatory transmitters is easy to rationalize. More surprisingly, many neurons in the motor system also show pacemaking activity, often rapid, including cerebellar Purkinje neurons that fire spontaneously at 20-100 Hz, as well as key neurons in the basal ganglia, including subthalamic nucleus neurons and globus pallidus neurons. Although the spontaneous rhythmic firing of pacemaking neurons is phenomenologically similar to cardiac pacemaking, the underlying ionic mechanism in most neurons is quite different than for cardiac pacemaking. Few spontaneously active neurons rely on HCN 'pacemaker' channels for their activity. Most commonly, a central element is 'persistent' sodium current, steady-state subthreshold current carried by the same voltage-dependent sodium channels that underlie fast action potentials. Persistent sodium current is a steeply voltage-dependent current with a midpoint near -60 mV, which results in regenerative spontaneous depolarization once it produces a net inward current when summed with all other background currents, often at voltages as negative as -70 mV. This 'engine' of pacemaking is present in almost all neurons and must be held in check in non-pacemaking neurons by sufficiently large competing outward currents from background potassium channels. The intrinsic propensity of neurons to fire spontaneously underlies key normal functions such as respiration and generates the complex background oscillatory circuits revealed in EEGs, but can also produce out-of-control oscillations of overall brain function in epilepsy, ataxia and tremor.
Collapse
Affiliation(s)
- Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Lucas-Romero J, Rivera-Arconada I, Lopez-Garcia JA. Noise or signal? Spontaneous activity of dorsal horn neurons: patterns and function in health and disease. Pflugers Arch 2024; 476:1171-1186. [PMID: 38822875 PMCID: PMC11271371 DOI: 10.1007/s00424-024-02971-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/10/2024] [Accepted: 05/05/2024] [Indexed: 06/03/2024]
Abstract
Spontaneous activity refers to the firing of action potentials by neurons in the absence of external stimulation. Initially considered an artifact or "noise" in the nervous system, it is now recognized as a potential feature of neural function. Spontaneous activity has been observed in various brain areas, in experimental preparations from different animal species, and in live animals and humans using non-invasive imaging techniques. In this review, we specifically focus on the spontaneous activity of dorsal horn neurons of the spinal cord. We use a historical perspective to set the basis for a novel classification of the different patterns of spontaneous activity exhibited by dorsal horn neurons. Then we examine the origins of this activity and propose a model circuit to explain how the activity is generated and transmitted to the dorsal horn. Finally, we discuss possible roles of this activity during development and during signal processing under physiological conditions and pain states. By analyzing recent studies on the spontaneous activity of dorsal horn neurons, we aim to shed light on its significance in sensory processing. Understanding the different patterns of activity, the origins of this activity, and the potential roles it may play, will contribute to our knowledge of sensory mechanisms, including pain, to facilitate the modeling of spinal circuits and hopefully to explore novel strategies for pain treatment.
Collapse
Affiliation(s)
- Javier Lucas-Romero
- Department of Systems Biology, University of Alcala, 28805, Madrid, Spain
- Department of Physical Therapy, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | | | - Jose Antonio Lopez-Garcia
- Department of Systems Biology, University of Alcala, 28805, Madrid, Spain.
- Departamento de Biologia de Sistemas, Edificio de Medicina, Universidad de Alcala, Ctra. Madrid-Barcelona, Km 33,600, 28805, Alcala de Henares, Madrid, Spain.
| |
Collapse
|
4
|
Yang YL, Lee HF, Chi CS, Tsai CR, Wu PY, Liu SN. Cerebellar atrophy in genetic epileptic encephalopathies: A cohort study and a systematic review. Seizure 2024; 120:41-48. [PMID: 38897163 DOI: 10.1016/j.seizure.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/29/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE To analyze cerebellar atrophy in genetic epileptic encephalopathies (EEs). METHODS This research included a retrospective cohort study conducted from January 2016 to December 2023 and a systematic review on cerebellar atrophy in genetic EEs. Pediatric individuals who were diagnosed with EEs based on electroclinical features, carried causative gene variants, and exhibited cerebellar atrophy were recruited. Electroclinical features, neuroimaging findings, and causative variants of eligible individuals were analyzed. RESULTS The cohort study showed 10 of 67 pediatric individuals (10/67; 15 %) who were diagnosed with genetic EEs had cerebellar atrophy; and 6 of the 10 individuals (6/10; 60 %) exhibited cerebellar signs. Diagnostic delay between the detection of cerebellar atrophy and the identification of genetic diagnosis existed in 6 individuals (6/10; 60 %) and the median duration was 4.4 years. A total of 32 genes, including 31 genes from the literature review and a newly identified SCN2A gene in this cohort, were reported associated with cerebellar atrophy in genetic EEs. Twenty-six genes (26/32; 81 %) accounted for cerebellar atrophy associated with other brain anomalies and 6 genes (6/32; 19 %) caused isolated cerebellar atrophy. Twenty-five genes (25/32; 78 %) showed late-onset cerebellar atrophy identified after the age of 1 year old. CONCLUSION Cerebellar atrophy is not uncommon in genetic EEs and may serve as an indicator for molecular diagnosis in clinical practice. To shorten the diagnostic delay, follow-up neuroimaging study is crucial because of high rate of clinico-radiological dissociation and late-onset cerebellar atrophy in this patient group.
Collapse
Affiliation(s)
- Yao-Lun Yang
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan
| | - Hsiu-Fen Lee
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, 145, Xingda Rd., Taichung 402, Taiwan.
| | - Ching-Shiang Chi
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan
| | - Chi-Ren Tsai
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan
| | - Pei-Yu Wu
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan
| | - Shu-Ning Liu
- Division of Pediatric Neurology, Children's Medical Center, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung 407, Taiwan
| |
Collapse
|
5
|
Brown SP, Lawson RJ, Moreno JD, Ransdell JL. A Reinterpretation of the Relationship between Persistent and Resurgent Sodium Currents. J Neurosci 2024; 44:e2396232024. [PMID: 38858080 PMCID: PMC11255426 DOI: 10.1523/jneurosci.2396-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/14/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
The resurgent sodium current (INaR) activates on membrane repolarization, such as during the downstroke of neuronal action potentials. Due to its unique activation properties, INaR is thought to drive high rates of repetitive neuronal firing. However, INaR is often studied in combination with the persistent or noninactivating portion of sodium currents (INaP). We used dynamic clamp to test how INaR and INaP individually affect repetitive firing in adult cerebellar Purkinje neurons from male and female mice. We learned INaR does not scale repetitive firing rates due to its rapid decay at subthreshold voltages and that subthreshold INaP is critical in regulating neuronal firing rate. Adjustments to the voltage-gated sodium conductance model used in these studies revealed INaP and INaR can be inversely scaled by adjusting occupancy in the slow-inactivated kinetic state. Together with additional dynamic clamp experiments, these data suggest the regulation of sodium channel slow inactivation can fine-tune INaP and Purkinje neuron repetitive firing rates.
Collapse
Affiliation(s)
- Samuel P Brown
- Department of Biology, Miami University, Oxford, Ohio 45056
| | - Ryan J Lawson
- Department of Biology, Miami University, Oxford, Ohio 45056
| | - Jonathan D Moreno
- Division of Cardiology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri 63130
| | | |
Collapse
|
6
|
Aman TK, Raman IM. Resurgent current in context: Insights from the structure and function of Na and K channels. Biophys J 2024; 123:1924-1941. [PMID: 38130058 PMCID: PMC11309984 DOI: 10.1016/j.bpj.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Discovered just over 25 years ago in cerebellar Purkinje neurons, resurgent Na current was originally described operationally as a component of voltage-gated Na current that flows upon repolarization from relatively depolarized potentials and speeds recovery from inactivation, increasing excitability. Its presence in many excitable cells and absence from others has raised questions regarding its biophysical and molecular mechanisms. Early studies proposed that Na channels capable of generating resurgent current are subject to a rapid open-channel block by an endogenous blocking protein, which binds upon depolarization and unblocks upon repolarization. Since the time that this mechanism was suggested, many physiological and structural studies of both Na and K channels have revealed aspects of gating and conformational states that provide insights into resurgent current. These include descriptions of domain movements for activation and inactivation, solution of cryo-EM structures with pore-blocking compounds, and identification of native blocking domains, proteins, and modulatory subunits. Such results not only allow the open-channel block hypothesis to be refined but also link it more clearly to research that preceded it. This review considers possible mechanisms for resurgent Na current in the context of earlier and later studies of ion channels and suggests a framework for future research.
Collapse
Affiliation(s)
- Teresa K Aman
- Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, Illinois.
| |
Collapse
|
7
|
Fernández Santoro EM, Karim A, Warnaar P, De Zeeuw CI, Badura A, Negrello M. Purkinje cell models: past, present and future. Front Comput Neurosci 2024; 18:1426653. [PMID: 39049990 PMCID: PMC11266113 DOI: 10.3389/fncom.2024.1426653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
The investigation of the dynamics of Purkinje cell (PC) activity is crucial to unravel the role of the cerebellum in motor control, learning and cognitive processes. Within the cerebellar cortex (CC), these neurons receive all the incoming sensory and motor information, transform it and generate the entire cerebellar output. The relatively homogenous and repetitive structure of the CC, common to all vertebrate species, suggests a single computation mechanism shared across all PCs. While PC models have been developed since the 70's, a comprehensive review of contemporary models is currently lacking. Here, we provide an overview of PC models, ranging from the ones focused on single cell intracellular PC dynamics, through complex models which include synaptic and extrasynaptic inputs. We review how PC models can reproduce physiological activity of the neuron, including firing patterns, current and multistable dynamics, plateau potentials, calcium signaling, intrinsic and synaptic plasticity and input/output computations. We consider models focusing both on somatic and on dendritic computations. Our review provides a critical performance analysis of PC models with respect to known physiological data. We expect our synthesis to be useful in guiding future development of computational models that capture real-life PC dynamics in the context of cerebellar computations.
Collapse
Affiliation(s)
| | - Arun Karim
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Pascal Warnaar
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | | | - Mario Negrello
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
8
|
Voerman S, Broersen R, Swagemakers SMA, De Zeeuw CI, van der Spek PJ. Plasticity mechanisms of genetically distinct Purkinje cells. Bioessays 2024; 46:e2400008. [PMID: 38697917 DOI: 10.1002/bies.202400008] [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: 01/12/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024]
Abstract
Despite its uniform appearance, the cerebellar cortex is highly heterogeneous in terms of structure, genetics and physiology. Purkinje cells (PCs), the principal and sole output neurons of the cerebellar cortex, can be categorized into multiple populations that differentially express molecular markers and display distinctive physiological features. Such features include action potential rate, but also their propensity for synaptic and intrinsic plasticity. However, the precise molecular and genetic factors that correlate with the differential physiological properties of PCs remain elusive. In this article, we provide a detailed overview of the cellular mechanisms that regulate PC activity and plasticity. We further perform a pathway analysis to highlight how molecular characteristics of specific PC populations may influence their physiology and plasticity mechanisms.
Collapse
Affiliation(s)
- Stijn Voerman
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Robin Broersen
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Sigrid M A Swagemakers
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Peter J van der Spek
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Brown SP, Lawson RJ, Moreno JD, Ransdell JL. A Reinterpretation of the Relationship Between Persistent and Resurgent Sodium Currents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.25.564042. [PMID: 38187680 PMCID: PMC10769191 DOI: 10.1101/2023.10.25.564042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The resurgent sodium current (INaR) activates on membrane repolarization, such as during the downstroke of neuronal action potentials. Due to its unique activation properties, INaR is thought to drive high rates of repetitive neuronal firing. However, INaR is often studied in combination with the persistent or non-inactivating portion of sodium currents (INaP). We used dynamic clamp to test how INaR and INaP individually affect repetitive firing in adult cerebellar Purkinje neurons from male and female mice. We learned INaR does not scale repetitive firing rates due to its rapid decay at subthreshold voltages, and that subthreshold INaP is critical in regulating neuronal firing rate. Adjustments to the Nav conductance model used in these studies revealed INaP and INaR can be inversely scaled by adjusting occupancy in the slow inactivated kinetic state. Together with additional dynamic clamp experiments, these data suggest the regulation of sodium channel slow inactivation can fine-tune INaP and Purkinje neuron repetitive firing rates.
Collapse
Affiliation(s)
| | - Ryan J. Lawson
- Department of Biology, Miami University, Oxford, OH 45056
| | - Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130
| | | |
Collapse
|
10
|
González-Tapia D, Vázquez-Hernández N, Urmeneta-Ortiz F, Navidad-Hernandez N, Lazo-Yepez M, Tejeda-Martínez A, Flores-Soto M, González-Burgos I. 3-Acetylpyridine-induced ataxic-like motor impairments are associated with plastic changes in the Purkinje cells of the rat cerebellum. Neurologia 2024; 39:408-416. [PMID: 38830720 DOI: 10.1016/j.nrleng.2021.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/18/2021] [Indexed: 06/05/2024] Open
Abstract
Ataxias are characterized by aberrant movement patterns closely related to cerebellar dysfunction. Purkinje cell axons are the sole outputs from the cerebellar cortex, and dysfunctional activity of Purkinje cells has been associated with ataxic movements. However, the synaptic characteristics of Purkinje cells in cases of ataxia are not yet well understood. The nicotinamide antagonist 3-acethylpyridine (3-AP) selectively destroys inferior olivary nucleus neurons so it is widely used to induce cerebellar ataxia. Five days after 3-AP treatment (65mg/kg) in adult male Sprague-Dawley rats, motor incoordination was revealed through BBB and Rotarod testing. In addition, in Purkinje cells from lobules V-VII of the cerebellar vermis studied by the Golgi method, the density of dendritic spines decreased, especially the thin and mushroom types. Western blot analysis showed a decrease in AMPA and PSD-95 content with an increase of the α-catenin protein, while GAD-67 and synaptophysin were unchanged. Findings suggest a limited capacity of Purkinje cells to acquire and consolidate afferent excitatory inputs and an aberrant, rigid profile in the movement-related output patterns of Purkinje neurons that likely contributes to the motor-related impairments characteristic of cerebellar ataxias.
Collapse
Affiliation(s)
- D González-Tapia
- Centro Universitario de Tlajomulco, Universidad de Guadalajara, Tlajomulco de Zúñiga, Jal., Mexico
| | - N Vázquez-Hernández
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - F Urmeneta-Ortiz
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - N Navidad-Hernandez
- Universidad Politécnica de la Zona Metropolitana de Guadalajara, Tlajomulco de Zúñiga, Jal., Mexico
| | - M Lazo-Yepez
- Universidad Politécnica de la Zona Metropolitana de Guadalajara, Tlajomulco de Zúñiga, Jal., Mexico
| | - A Tejeda-Martínez
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - M Flores-Soto
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico
| | - I González-Burgos
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jal., Mexico.
| |
Collapse
|
11
|
Wu J, Shindo Y, Hotta K, Vu CQ, Lu K, Wazawa T, Nagai T, Oka K. Calcium-induced upregulation of energy metabolism heats neurons during neural activity. Biochem Biophys Res Commun 2024; 708:149799. [PMID: 38522401 DOI: 10.1016/j.bbrc.2024.149799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/13/2024] [Accepted: 03/16/2024] [Indexed: 03/26/2024]
Abstract
Cellular temperature affects every biochemical reaction, underscoring its critical role in cellular functions. In neurons, temperature not only modulates neurotransmission but is also a key determinant of neurodegenerative diseases. Considering that the brain consumes a disproportionately high amount of energy relative to its weight, neural circuits likely generate a lot of heat, which can increase cytosolic temperature. However, the changes in temperature within neurons and the mechanisms of heat generation during neural excitation remain unclear. In this study, we achieved simultaneous imaging of Ca2+ and temperature using the genetically encoded indicators, B-GECO and B-gTEMP. We then compared the spatiotemporal distributions of Ca2+ responses and temperature. Following neural excitation induced by veratridine, an activator of the voltage-gated Na+ channel, we observed an approximately 2 °C increase in cytosolic temperature occurring 30 s after the Ca2+ response. The temperature elevation was observed in the non-nuclear region, while Ca2+ increased throughout the cell body. Moreover, this temperature increase was suppressed under Ca2+-free conditions and by inhibitors of ATP synthesis. These results indicate that Ca2+-induced upregulation of energy metabolism serves as the heat source during neural excitation.
Collapse
Affiliation(s)
- Jiayang Wu
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Yutaka Shindo
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan; School of Frontier Engineering, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Kohji Hotta
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Cong Quang Vu
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Kai Lu
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Tetsuichi Wazawa
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Takeharu Nagai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Kotaro Oka
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan; School of Frontier Engineering, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan; Waseda Research Institute for Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan.
| |
Collapse
|
12
|
Beau M, Herzfeld DJ, Naveros F, Hemelt ME, D’Agostino F, Oostland M, Sánchez-López A, Chung YY, Michael Maibach, Kyranakis S, Stabb HN, Martínez Lopera MG, Lajko A, Zedler M, Ohmae S, Hall NJ, Clark BA, Cohen D, Lisberger SG, Kostadinov D, Hull C, Häusser M, Medina JF. A deep-learning strategy to identify cell types across species from high-density extracellular recordings. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.577845. [PMID: 38352514 PMCID: PMC10862837 DOI: 10.1101/2024.01.30.577845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
High-density probes allow electrophysiological recordings from many neurons simultaneously across entire brain circuits but don't reveal cell type. Here, we develop a strategy to identify cell types from extracellular recordings in awake animals, revealing the computational roles of neurons with distinct functional, molecular, and anatomical properties. We combine optogenetic activation and pharmacology using the cerebellum as a testbed to generate a curated ground-truth library of electrophysiological properties for Purkinje cells, molecular layer interneurons, Golgi cells, and mossy fibers. We train a semi-supervised deep-learning classifier that predicts cell types with greater than 95% accuracy based on waveform, discharge statistics, and layer of the recorded neuron. The classifier's predictions agree with expert classification on recordings using different probes, in different laboratories, from functionally distinct cerebellar regions, and across animal species. Our classifier extends the power of modern dynamical systems analyses by revealing the unique contributions of simultaneously-recorded cell types during behavior.
Collapse
Affiliation(s)
- Maxime Beau
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - David J. Herzfeld
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Francisco Naveros
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Computer Engineering, Automation and Robotics, Research Centre for Information and Communication Technologies, University of Granada, Granada, Spain
| | - Marie E. Hemelt
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Federico D’Agostino
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Marlies Oostland
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Young Yoon Chung
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Michael Maibach
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Stephen Kyranakis
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Hannah N. Stabb
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Agoston Lajko
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Marie Zedler
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Shogo Ohmae
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Nathan J. Hall
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Beverley A. Clark
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Dana Cohen
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Dimitar Kostadinov
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Centre for Developmental Neurobiology, King’s College London, London, UK
| | - Court Hull
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Michael Häusser
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Javier F. Medina
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
13
|
Gu J, Rollo B, Berecki G, Petrou S, Kwan P, Sumer H, Cromer B. Generation of a stably transfected mouse embryonic stem cell line for inducible differentiation to excitatory neurons. Exp Cell Res 2024; 435:113902. [PMID: 38145818 DOI: 10.1016/j.yexcr.2023.113902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 12/27/2023]
Abstract
In vitro differentiation of stem cells into various cell lineages is valuable in developmental studies and an important source of cells for modelling physiology and pathology, particularly for complex tissues such as the brain. Conventional protocols for in vitro neuronal differentiation often suffer from complicated procedures, high variability and low reproducibility. Over the last decade, the identification of cell fate-determining transcription factors has provided new tools for cellular studies in neuroscience and enabled rapid differentiation driven by ectopic transcription factor expression. As a proneural transcription factor, Neurogenin 2 (Ngn2) expression alone is sufficient to trigger rapid and robust neurogenesis from pluripotent cells. Here, we established a stable cell line, by piggyBac (PB) transposition, that conditionally expresses Ngn2 for generation of excitatory neurons from mouse embryonic stem cells (ESCs) using an all-in-one PB construct. Our results indicate that Ngn2-induced excitatory neurons have mature and functional characteristics consistent with previous studies using conventional differentiation methods. This approach provides an all-in-one PB construct for rapid and high copy number gene delivery of dox-inducible transcription factors to induce differentiation. This approach is a valuable in vitro cell model for disease modeling, drug screening and cell therapy.
Collapse
Affiliation(s)
- Jinchao Gu
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Australia; Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Melbourne, Australia
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Melbourne, Australia
| | - Geza Berecki
- Ion Channels and Human Diseases Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Steven Petrou
- Ion Channels and Human Diseases Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Melbourne, Australia
| | - Huseyin Sumer
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Australia.
| | - Brett Cromer
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Australia.
| |
Collapse
|
14
|
Orfali R, Alwatban AZ, Orfali RS, Lau L, Chea N, Alotaibi AM, Nam YW, Zhang M. Oxidative stress and ion channels in neurodegenerative diseases. Front Physiol 2024; 15:1320086. [PMID: 38348223 PMCID: PMC10859863 DOI: 10.3389/fphys.2024.1320086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
Numerous neurodegenerative diseases result from altered ion channel function and mutations. The intracellular redox status can significantly alter the gating characteristics of ion channels. Abundant neurodegenerative diseases associated with oxidative stress have been documented, including Parkinson's, Alzheimer's, spinocerebellar ataxia, amyotrophic lateral sclerosis, and Huntington's disease. Reactive oxygen and nitrogen species compounds trigger posttranslational alterations that target specific sites within the subunits responsible for channel assembly. These alterations include the adjustment of cysteine residues through redox reactions induced by reactive oxygen species (ROS), nitration, and S-nitrosylation assisted by nitric oxide of tyrosine residues through peroxynitrite. Several ion channels have been directly investigated for their functional responses to oxidizing agents and oxidative stress. This review primarily explores the relationship and potential links between oxidative stress and ion channels in neurodegenerative conditions, such as cerebellar ataxias and Parkinson's disease. The potential correlation between oxidative stress and ion channels could hold promise for developing innovative therapies for common neurodegenerative diseases.
Collapse
Affiliation(s)
- Razan Orfali
- Neuroscience Research Department, Research Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Adnan Z. Alwatban
- Neuroscience Research Department, Research Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Liz Lau
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Noble Chea
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Abdullah M. Alotaibi
- Neuroscience Research Department, Research Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| |
Collapse
|
15
|
Wu S, Wardak A, Khan MM, Chen CH, Regehr WG. Implications of variable synaptic weights for rate and temporal coding of cerebellar outputs. eLife 2024; 13:e89095. [PMID: 38241596 PMCID: PMC10798666 DOI: 10.7554/elife.89095] [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: 05/03/2023] [Accepted: 12/27/2023] [Indexed: 01/21/2024] Open
Abstract
Purkinje cell (PC) synapses onto cerebellar nuclei (CbN) neurons allow signals from the cerebellar cortex to influence the rest of the brain. PCs are inhibitory neurons that spontaneously fire at high rates, and many PC inputs are thought to converge onto each CbN neuron to suppress its firing. It has been proposed that PCs convey information using a rate code, a synchrony and timing code, or both. The influence of PCs on CbN neuron firing was primarily examined for the combined effects of many PC inputs with comparable strengths, and the influence of individual PC inputs has not been extensively studied. Here, we find that single PC to CbN synapses are highly variable in size, and using dynamic clamp and modeling we reveal that this has important implications for PC-CbN transmission. Individual PC inputs regulate both the rate and timing of CbN firing. Large PC inputs strongly influence CbN firing rates and transiently eliminate CbN firing for several milliseconds. Remarkably, the refractory period of PCs leads to a brief elevation of CbN firing prior to suppression. Thus, individual PC-CbN synapses are suited to concurrently convey rate codes and generate precisely timed responses in CbN neurons. Either synchronous firing or synchronous pauses of PCs promote CbN neuron firing on rapid time scales for nonuniform inputs, but less effectively than for uniform inputs. This is a secondary consequence of variable input sizes elevating the baseline firing rates of CbN neurons by increasing the variability of the inhibitory conductance. These findings may generalize to other brain regions with highly variable inhibitory synapse sizes.
Collapse
Affiliation(s)
- Shuting Wu
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Asem Wardak
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mehak M Khan
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Christopher H Chen
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of MedicineHersheyUnited States
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
16
|
Atamian A, Birtele M, Hosseini N, Nguyen T, Seth A, Del Dosso A, Paul S, Tedeschi N, Taylor R, Coba MP, Samarasinghe R, Lois C, Quadrato G. Human cerebellar organoids with functional Purkinje cells. Cell Stem Cell 2024; 31:39-51.e6. [PMID: 38181749 PMCID: PMC11417151 DOI: 10.1016/j.stem.2023.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/30/2023] [Accepted: 11/30/2023] [Indexed: 01/07/2024]
Abstract
Research on human cerebellar development and disease has been hampered by the need for a human cell-based system that recapitulates the human cerebellum's cellular diversity and functional features. Here, we report a human organoid model (human cerebellar organoids [hCerOs]) capable of developing the complex cellular diversity of the fetal cerebellum, including a human-specific rhombic lip progenitor population that have never been generated in vitro prior to this study. 2-month-old hCerOs form distinct cytoarchitectural features, including laminar organized layering, and create functional connections between inhibitory and excitatory neurons that display coordinated network activity. Long-term culture of hCerOs allows healthy survival and maturation of Purkinje cells that display molecular and electrophysiological hallmarks of their in vivo counterparts, addressing a long-standing challenge in the field. This study therefore provides a physiologically relevant, all-human model system to elucidate the cell-type-specific mechanisms governing cerebellar development and disease.
Collapse
Affiliation(s)
- Alexander Atamian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Marcella Birtele
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Negar Hosseini
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tuan Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Anoothi Seth
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ashley Del Dosso
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sandeep Paul
- Spatial Genomics, 145 Vista Avenue Suite 111, Pasadena, CA 91107, USA
| | - Neil Tedeschi
- Spatial Genomics, 145 Vista Avenue Suite 111, Pasadena, CA 91107, USA
| | - Ryan Taylor
- Spatial Genomics, 145 Vista Avenue Suite 111, Pasadena, CA 91107, USA
| | - Marcelo P Coba
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA 90033, USA
| | - Ranmal Samarasinghe
- Department of Clinical Neurophysiology and Neurology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
17
|
Raman IM. The Hodgkin-Huxley-Katz Prize Lecture: A Markov model with permeation-dependent gating that accounts for resurgent current of voltage-gated Na channels. J Physiol 2023; 601:5147-5164. [PMID: 37837315 PMCID: PMC10913027 DOI: 10.1113/jp285166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/20/2023] [Indexed: 10/16/2023] Open
Abstract
Many neurons that fire high-frequency action potentials express specialized voltage-gated Na channel complexes that not only conduct transient current upon depolarization, but also pass resurgent current upon repolarization. The resurgent current is associated with recovery of transient current, even at moderately negative potentials where fast inactivation is usually absorbing. The combined results of many experimental studies have led to the hypothesis that resurgent current flows upon repolarization when an endogenous blocking protein that occludes open channels at depolarized potentials is expelled by inwardly permeating Na ions. Additional observations have suggested that the position of the voltage sensor of domain IV regulates the affinity of the channel for the putative blocker. To test the effectiveness of a kinetic scheme incorporating these features, here we develop and justify a Markov model with states grounded in known Na channel conformations. Simulations were designed to investigate whether including a permeation-dependent unblocking rate constant and two open-blocked states, superimposed on conformations and voltage-sensitive movements present in all voltage-gated Na channels, is sufficient to account for the unusual gating of channels with a resurgent component. Optimizing rate constant parameters against a wide range of experimental data from cerebellar Purkinje cells demonstrates that a kinetic scheme for Na channels incorporating the novel aspects of a permeation-dependent unblock, as well as distinct high- and low-affinity blocked states, reproduces all the attributes of experimentally recorded Na currents in a physiologically plausible manner.
Collapse
Affiliation(s)
- Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| |
Collapse
|
18
|
Pilotto F, Douthwaite C, Diab R, Ye X, Al Qassab Z, Tietje C, Mounassir M, Odriozola A, Thapa A, Buijsen RAM, Lagache S, Uldry AC, Heller M, Müller S, van Roon-Mom WMC, Zuber B, Liebscher S, Saxena S. Early molecular layer interneuron hyperactivity triggers Purkinje neuron degeneration in SCA1. Neuron 2023; 111:2523-2543.e10. [PMID: 37321222 PMCID: PMC10431915 DOI: 10.1016/j.neuron.2023.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/17/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Toxic proteinaceous deposits and alterations in excitability and activity levels characterize vulnerable neuronal populations in neurodegenerative diseases. Using in vivo two-photon imaging in behaving spinocerebellar ataxia type 1 (Sca1) mice, wherein Purkinje neurons (PNs) degenerate, we identify an inhibitory circuit element (molecular layer interneurons [MLINs]) that becomes prematurely hyperexcitable, compromising sensorimotor signals in the cerebellum at early stages. Mutant MLINs express abnormally elevated parvalbumin, harbor high excitatory-to-inhibitory synaptic density, and display more numerous synaptic connections on PNs, indicating an excitation/inhibition imbalance. Chemogenetic inhibition of hyperexcitable MLINs normalizes parvalbumin expression and restores calcium signaling in Sca1 PNs. Chronic inhibition of mutant MLINs delayed PN degeneration, reduced pathology, and ameliorated motor deficits in Sca1 mice. Conserved proteomic signature of Sca1 MLINs, shared with human SCA1 interneurons, involved the higher expression of FRRS1L, implicated in AMPA receptor trafficking. We thus propose that circuit-level deficits upstream of PNs are one of the main disease triggers in SCA1.
Collapse
Affiliation(s)
- Federica Pilotto
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Christopher Douthwaite
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Rim Diab
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - XiaoQian Ye
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Zahraa Al Qassab
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Christoph Tietje
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Meriem Mounassir
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | | | - Aishwarya Thapa
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sophie Lagache
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stefan Müller
- Flow Cytometry and Cell sorting, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Benoît Zuber
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; University Hospital Cologne, Deptartment of Neurology, Cologne, Germany.
| | - Smita Saxena
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
Herzog R, Bolte C, Radecke JO, von Möller K, Lencer R, Tzvi E, Münchau A, Bäumer T, Weissbach A. Neuronavigated Cerebellar 50 Hz tACS: Attenuation of Stimulation Effects by Motor Sequence Learning. Biomedicines 2023; 11:2218. [PMID: 37626715 PMCID: PMC10452137 DOI: 10.3390/biomedicines11082218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Cerebellar transcranial alternating current stimulation (tACS) is an emerging non-invasive technique that induces electric fields to modulate cerebellar function. Although the effect of cortical tACS seems to be state-dependent, the impact of concurrent motor activation and the duration of stimulation on the effects of cerebellar tACS has not yet been examined. In our study, 20 healthy subjects received neuronavigated 50 Hz cerebellar tACS for 40 s or 20 min, each during performance using a motor sequence learning task (MSL) and at rest. We measured the motor evoked potential (MEP) before and at two time points after tACS application to assess corticospinal excitability. Additionally, we investigated the online effect of tACS on MSL. Individual electric field simulations were computed to evaluate the distribution of electric fields, showing a focal electric field in the right cerebellar hemisphere with the highest intensities in lobe VIIb, VIII and IX. Corticospinal excitability was only increased after tACS was applied for 40 s or 20 min at rest, and motor activation during tACS (MSL) cancelled this effect. In addition, performance was better (shorter reaction times) for the learned sequences after 20 min of tACS, indicating more pronounced learning under 20 min of tACS compared to tACS applied only in the first 40 s.
Collapse
Affiliation(s)
- Rebecca Herzog
- Institute of Systems Motor Science, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (R.H.); (C.B.)
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Department of Neurology, University Hospital Schleswig Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Christina Bolte
- Institute of Systems Motor Science, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (R.H.); (C.B.)
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Jan-Ole Radecke
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Kathinka von Möller
- Institute of Systems Motor Science, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (R.H.); (C.B.)
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Rebekka Lencer
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Elinor Tzvi
- Department of Neurology, Leipzig University, Liebigstraße 20, 04103 Leipzig, Germany
- Syte Institute, Hohe Bleichen 8, 20354 Hamburg, Germany
| | - Alexander Münchau
- Institute of Systems Motor Science, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (R.H.); (C.B.)
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Tobias Bäumer
- Institute of Systems Motor Science, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (R.H.); (C.B.)
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Anne Weissbach
- Institute of Systems Motor Science, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (R.H.); (C.B.)
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| |
Collapse
|
20
|
Buijsen RAM, Hu M, Sáez-González M, Notopoulou S, Mina E, Koning W, Gardiner SL, van der Graaf LM, Daoutsali E, Pepers BA, Mei H, van Dis V, Frimat JP, van den Maagdenberg AMJM, Petrakis S, van Roon-Mom WMC. Spinocerebellar Ataxia Type 1 Characteristics in Patient-Derived Fibroblast and iPSC-Derived Neuronal Cultures. Mov Disord 2023; 38:1428-1442. [PMID: 37278528 DOI: 10.1002/mds.29446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/23/2023] [Accepted: 04/20/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by a polyglutamine expansion in the ataxin-1 protein resulting in neuropathology including mutant ataxin-1 protein aggregation, aberrant neurodevelopment, and mitochondrial dysfunction. OBJECTIVES Identify SCA1-relevant phenotypes in patient-specific fibroblasts and SCA1 induced pluripotent stem cells (iPSCs) neuronal cultures. METHODS SCA1 iPSCs were generated and differentiated into neuronal cultures. Protein aggregation and neuronal morphology were evaluated using fluorescent microscopy. Mitochondrial respiration was measured using the Seahorse Analyzer. The multi-electrode array (MEA) was used to identify network activity. Finally, gene expression changes were studied using RNA-seq to identify disease-specific mechanisms. RESULTS Bioenergetics deficits in patient-derived fibroblasts and SCA1 neuronal cultures showed altered oxygen consumption rate, suggesting involvement of mitochondrial dysfunction in SCA1. In SCA1 hiPSC-derived neuronal cells, nuclear and cytoplasmic aggregates were identified similar in localization as aggregates in SCA1 postmortem brain tissue. SCA1 hiPSC-derived neuronal cells showed reduced dendrite length and number of branching points while MEA recordings identified delayed development in network activity in SCA1 hiPSC-derived neuronal cells. Transcriptome analysis identified 1050 differentially expressed genes in SCA1 hiPSC-derived neuronal cells associated with synapse organization and neuron projection guidance, where a subgroup of 151 genes was highly associated with SCA1 phenotypes and linked to SCA1 relevant signaling pathways. CONCLUSIONS Patient-derived cells recapitulate key pathological features of SCA1 pathogenesis providing a valuable tool for the identification of novel disease-specific processes. This model can be used for high throughput screenings to identify compounds, which may prevent or rescue neurodegeneration in this devastating disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Michel Hu
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Maria Sáez-González
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sofia Notopoulou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Eleni Mina
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Winette Koning
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sarah L Gardiner
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Elena Daoutsali
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Barry A Pepers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Vera van Dis
- Department of Pathology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Pathology, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Jean-Philippe Frimat
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Spyros Petrakis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
21
|
Huang H, Shakkottai VG. Targeting Ion Channels and Purkinje Neuron Intrinsic Membrane Excitability as a Therapeutic Strategy for Cerebellar Ataxia. Life (Basel) 2023; 13:1350. [PMID: 37374132 DOI: 10.3390/life13061350] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
In degenerative neurological disorders such as Parkinson's disease, a convergence of widely varying insults results in a loss of dopaminergic neurons and, thus, the motor symptoms of the disease. Dopamine replacement therapy with agents such as levodopa is a mainstay of therapy. Cerebellar ataxias, a heterogeneous group of currently untreatable conditions, have not been identified to have a shared physiology that is a target of therapy. In this review, we propose that perturbations in cerebellar Purkinje neuron intrinsic membrane excitability, a result of ion channel dysregulation, is a common pathophysiologic mechanism that drives motor impairment and vulnerability to degeneration in cerebellar ataxias of widely differing genetic etiologies. We further propose that treatments aimed at restoring Purkinje neuron intrinsic membrane excitability have the potential to be a shared therapy in cerebellar ataxia akin to levodopa for Parkinson's disease.
Collapse
Affiliation(s)
- Haoran Huang
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Vikram G Shakkottai
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
22
|
Lobule-Related Action Potential Shape- and History-Dependent Current Integration in Purkinje Cells of Adult and Developing Mice. Cells 2023; 12:cells12040623. [PMID: 36831290 PMCID: PMC9953991 DOI: 10.3390/cells12040623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Purkinje cells (PCs) are the principal cells of the cerebellar cortex and form a central element in the modular organization of the cerebellum. Differentiation of PCs based on gene expression profiles revealed two subpopulations with distinct connectivity, action potential firing and learning-induced activity changes. However, which basal cell physiological features underlie the differences between these subpopulations and to what extent they integrate input differentially remains largely unclear. Here, we investigate the cellular electrophysiological properties of PC subpopulation in adult and juvenile mice. We found that multiple fundamental cell physiological properties, including membrane resistance and various aspects of the action potential shape, differ between PCs from anterior and nodular lobules. Moreover, the two PC subpopulations also differed in the integration of negative and positive current steps as well as in size of the hyperpolarization-activated current. A comparative analysis in juvenile mice confirmed that most of these lobule-specific differences are already present at pre-weaning ages. Finally, we found that current integration in PCs is input history-dependent for both positive and negative currents, but this is not a distinctive feature between anterior and nodular PCs. Our results support the concept of a fundamental differentiation of PCs subpopulations in terms of cell physiological properties and current integration, yet reveals that history-dependent input processing is consistent across PC subtypes.
Collapse
|
23
|
Lyu S, Xing H, Liu Y, Girdhar P, Yokoi F, Li Y. Further Studies on the Role of BTBD9 in the Cerebellum, Sleep-like Behaviors and the Restless Legs Syndrome. Neuroscience 2022; 505:78-90. [PMID: 36244636 PMCID: PMC10367443 DOI: 10.1016/j.neuroscience.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/15/2022] [Accepted: 10/09/2022] [Indexed: 11/26/2022]
Abstract
Genetic analyses have linked BTBD9 to restless legs syndrome (RLS) and sleep regulation. Btbd9 knockout mice show RLS-like motor restlessness. Previously, we found hyperactivity of cerebellar Purkinje cells (PCs) in Btbd9 knockout mice, which may contribute to the motor restlessness observed. However, underlying mechanisms for PC hyperactivity in Btbd9 knockout mice are unknown. Here, we used dissociated PC recording, brain slice recording and western blot to address this question. Our dissociated recording shows that knockout PCs had increased TEA-sensitive, Ca2+-dependent K+ currents. Applying antagonist to large conductance Ca2+-activated K+ (BK) channels further isolated the increased current as BK current. Consistently, we found increased amplitude of afterhyperpolarization and elevated BK protein levels in the knockout mice. Dissociated recording also shows a decrease in TEA-insensitive, Ca2+-dependent K+ currents. The result is consistent with reduced amplitude of tail currents, mainly composed of small conductance Ca2+-activated K+ (SK) currents, in slice recording. Our results suggest that BK and SK channels may be responsible for the hyperactivity of knockout PCs. Recently, BTBD9 protein was shown to associate with SYNGAP1 protein. We found a decreased cerebellar level of SYNGAP1 in Btbd9 knockout mice. However, Syngap1 heterozygous knockout mice showed nocturnal, instead of diurnal, motor restlessness. Our results suggest that SYNGAP1 deficiency may not contribute directly to the RLS-like motor restlessness observed in Btbd9 knockout mice. Finally, we found that PC-specific Btbd9 knockout mice exhibited deficits in motor coordination and balance similar to Btbd9 knockout mice, suggesting that the motor effect of BTBD9 in PCs is cell-autonomous.
Collapse
Affiliation(s)
- Shangru Lyu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuning Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pallavi Girdhar
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
24
|
van der Heijden ME, Brown AM, Sillitoe RV. Influence of data sampling methods on the representation of neural spiking activity in vivo. iScience 2022; 25:105429. [PMID: 36388953 PMCID: PMC9641233 DOI: 10.1016/j.isci.2022.105429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/06/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
In vivo single-unit recordings distinguish the basal spiking properties of neurons in different experimental settings and disease states. Here, we examined over 300 spike trains recorded from Purkinje cells and cerebellar nuclei neurons to test whether data sampling approaches influence the extraction of rich descriptors of firing properties. Our analyses included neurons recorded in awake and anesthetized control mice, and disease models of ataxia, dystonia, and tremor. We find that recording duration circumscribes overall representations of firing rate and pattern. Notably, shorter recording durations skew estimates for global firing rate variability toward lower values. We also find that only some populations of neurons in the same mouse are more similar to each other than to neurons recorded in different mice. These data reveal that recording duration and approach are primary considerations when interpreting task-independent single neuron firing properties. If not accounted for, group differences may be concealed or exaggerated.
Collapse
Affiliation(s)
- Meike E. van der Heijden
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Amanda M. Brown
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Roy V. Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
25
|
Thouta S, Waldbrook MG, Lin S, Mahadevan A, Mezeyova J, Soriano M, Versi P, Goodchild SJ, Parrish RR. Pharmacological determination of the fractional block of Nav channels required to impair neuronal excitability and ex vivo seizures. Front Cell Neurosci 2022; 16:964691. [PMID: 36246527 PMCID: PMC9557217 DOI: 10.3389/fncel.2022.964691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Voltage-gated sodium channels (Nav) are essential for the initiation and propagation of action potentials in neurons. Of the nine human channel subtypes, Nav1.1, Nav1.2 and Nav1.6 are prominently expressed in the adult central nervous system (CNS). All three of these sodium channel subtypes are sensitive to block by the neurotoxin tetrodotoxin (TTX), with TTX being almost equipotent on all three subtypes. In the present study we have used TTX to determine the fractional block of Nav channels required to impair action potential firing in pyramidal neurons and reduce network seizure-like activity. Using automated patch-clamp electrophysiology, we first determined the IC50s of TTX on mouse Nav1.1, Nav1.2 and Nav1.6 channels expressed in HEK cells, demonstrating this to be consistent with previously published data on human orthologs. We then compared this data to the potency of block of Nav current measured in pyramidal neurons from neocortical brain slices. Interestingly, we found that it requires nearly 10-fold greater concentration of TTX over the IC50 to induce significant block of action potentials using a current-step protocol. In contrast, concentrations near the IC50 resulted in a significant reduction in AP firing and increase in rheobase using a ramp protocol. Surprisingly, a 20% reduction in action potential generation observed with 3 nM TTX resulted in significant block of seizure-like activity in the 0 Mg2+ model of epilepsy. Additionally, we found that approximately 50% block in pyramidal cell intrinsic excitability is sufficient to completely block all seizure-like events. Furthermore, we also show that the anticonvulsant drug phenytoin blocked seizure-like events in a manner similar to TTX. These data serve as a critical starting point in understanding how fractional block of Nav channels affect intrinsic neuronal excitability and seizure-like activity. It further suggests that seizures can be controlled without significantly compromising intrinsic neuronal activity and determines the required fold over IC50 for novel and clinically relevant Nav channel blockers to produce efficacy and limit side effects.
Collapse
Affiliation(s)
- Samrat Thouta
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Matthew G. Waldbrook
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Sophia Lin
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Arjun Mahadevan
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Janette Mezeyova
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Maegan Soriano
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Pareesa Versi
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - Samuel J. Goodchild
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - R. Ryley Parrish
- Department of Cellular and Molecular Biology, Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| |
Collapse
|
26
|
Herzog R, Berger TM, Pauly MG, Xue H, Rueckert E, Münchau A, Bäumer T, Weissbach A. Cerebellar transcranial current stimulation – An intraindividual comparison of different techniques. Front Neurosci 2022; 16:987472. [PMID: 36188449 PMCID: PMC9521312 DOI: 10.3389/fnins.2022.987472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Transcranial current stimulation (tCS) techniques have been shown to induce cortical plasticity. As an important relay in the motor system, the cerebellum is an interesting target for plasticity induction using tCS, aiming to modulate its excitability and connectivity. However, until now it remains unclear, which is the most effective tCS method for inducing plasticity in the cerebellum. Thus, in this study, the effects of anodal transcranial direct current stimulation (tDCS), 50 Hz transcranial alternating current stimulation (50 Hz tACS), and high frequency transcranial random noise stimulation (tRNS) were compared with sham stimulation in 20 healthy subjects in a within-subject design. tCS was applied targeting the cerebellar lobe VIIIA using neuronavigation. We measured corticospinal excitability, short-interval intracortical inhibition (SICI), short-latency afferent inhibition (SAI), and cerebellar brain inhibition (CBI) and performed a sensor-based movement analysis at baseline and three times after the intervention (post1 = 15 min; post2 = 55 min; post3 = 95 min). Corticospinal excitability increased following cerebellar tACS and tRNS compared to sham stimulation. This effect was most pronounced directly after stimulation but lasted for at least 55 min after tACS. Cortico-cortical and cerebello-cortical conditioning protocols, as well as sensor-based movement analyses, did not change. Our findings suggest that cerebellar 50 Hz tACS is the most effective protocol to change corticospinal excitability.
Collapse
Affiliation(s)
- Rebecca Herzog
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | - Till M. Berger
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Martje G. Pauly
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Honghu Xue
- Institute for Robotics and Cognitive Systems, University of Lübeck, Lübeck, Germany
| | | | - Alexander Münchau
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Tobias Bäumer
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Anne Weissbach
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- *Correspondence: Anne Weissbach,
| |
Collapse
|
27
|
Chang HHV, Cook AA, Watt AJ, Cullen KE. Loss of Flocculus Purkinje Cell Firing Precision Leads to Impaired Gaze Stabilization in a Mouse Model of Spinocerebellar Ataxia Type 6 (SCA6). Cells 2022; 11:cells11172739. [PMID: 36078147 PMCID: PMC9454745 DOI: 10.3390/cells11172739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Spinocerebellar Ataxia Type 6 (SCA6) is a mid-life onset neurodegenerative disease characterized by progressive ataxia, dysarthria, and eye movement impairment. This autosomal dominant disease is caused by the expansion of a CAG repeat tract in the CACNA1A gene that encodes the α1A subunit of the P/Q type voltage-gated Ca2+ channel. Mouse models of SCA6 demonstrate impaired locomotive function and reduced firing precision of cerebellar Purkinje in the anterior vermis. Here, to further assess deficits in other cerebellar-dependent behaviors, we characterized the oculomotor phenotype of a knock-in mouse model with hyper-expanded polyQ repeats (SCA684Q). We found a reduction in the efficacy of the vestibulo-ocular reflex (VOR) and optokinetic reflex (OKR) in SCA6 mutant mice, without a change in phase, compared to their litter-matched controls. Additionally, VOR motor learning was significantly impaired in SCA684Q mice. Given that the floccular lobe of the cerebellum plays a vital role in the generation of OKR and VOR calibration and motor learning, we investigated the firing behavior and morphology of floccular cerebellar Purkinje cells. Overall, we found a reduction in the firing precision of floccular lobe Purkinje cells but no morphological difference between SCA684Q and wild-type mice. Taken together, our findings establish that gaze stabilization and motor learning are impaired in SCA684Q mice and suggest that altered cerebellar output contributes to these deficits.
Collapse
Affiliation(s)
| | - Anna A. Cook
- Department of Biology, McGill University, Montreal, QC H3G 0B1, Canada
| | - Alanna J. Watt
- Department of Biology, McGill University, Montreal, QC H3G 0B1, Canada
| | - Kathleen E. Cullen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
- Correspondence:
| |
Collapse
|
28
|
Abstract
The cerebellar cortex is an important system for relating neural circuits and learning. Its promise reflects the longstanding idea that it contains simple, repeated circuit modules with only a few cell types and a single plasticity mechanism that mediates learning according to classical Marr-Albus models. However, emerging data have revealed surprising diversity in neuron types, synaptic connections, and plasticity mechanisms, both locally and regionally within the cerebellar cortex. In light of these findings, it is not surprising that attempts to generate a holistic model of cerebellar learning across different behaviors have not been successful. While the cerebellum remains an ideal system for linking neuronal function with behavior, it is necessary to update the cerebellar circuit framework to achieve its great promise. In this review, we highlight recent advances in our understanding of cerebellar-cortical cell types, synaptic connections, signaling mechanisms, and forms of plasticity that enrich cerebellar processing.
Collapse
Affiliation(s)
- Court Hull
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA;
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
29
|
Srinivasan SR, Huang H, Chang WC, Nasburg JA, Nguyen HM, Strassmaier T, Wulff H, Shakkottai VG. Discovery of Novel Activators of Large-Conductance Calcium-Activated Potassium Channels for the Treatment of Cerebellar Ataxia. Mol Pharmacol 2022; 102:438-449. [PMID: 35489717 PMCID: PMC9341255 DOI: 10.1124/molpharm.121.000478] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/07/2022] [Indexed: 01/11/2023] Open
Abstract
Impaired cerebellar Purkinje neuron firing resulting from reduced expression of large-conductance calcium-activated potassium (BK) channels is a consistent feature in models of inherited neurodegenerative spinocerebellar ataxia (SCA). Restoring BK channel expression improves motor function and delays cerebellar degeneration, indicating that BK channels are an attractive therapeutic target. Current BK channel activators lack specificity and potency and are therefore poor templates for future drug development. We implemented an automated patch clamp platform for high-throughput drug discovery of BK channel activators using the Nanion SyncroPatch 384PE system. We screened over 15,000 compounds for their ability to increase BK channel current amplitude under conditions of lower intracellular calcium that is present in disease. We identified several novel BK channel activators that were then retested on the SyncroPatch 384PE to generate concentration-response curves (CRCs). Compounds with favorable CRCs were subsequently tested for their ability to improve irregular cerebellar Purkinje neuron spiking, characteristic of BK channel dysfunction in SCA1 mice. We identified a novel BK channel activator, 4-chloro-N-(5-chloro-2-cyanophenyl)-3-(trifluoromethyl)benzene-1-sulfonamide (herein renamed BK-20), that exhibited a more potent half-maximal activation of BK current (pAC50 = 4.64) than NS-1619 (pAC50 = 3.7) at a free internal calcium concentration of 270 nM in a heterologous expression system and improved spiking regularity in SCA1 Purkinje neurons. BK-20 had no activity on small-conductance calcium-activated potassium (SK)1-3 channels but interestingly was a potent blocker of the T-type calcium channel, Cav3.1 (IC50 = 1.05 μM). Our work describes both a novel compound for further drug development in disorders with irregular Purkinje spiking and a unique platform for drug discovery in degenerative ataxias. SIGNIFICANCE STATEMENT: Motor impairment associated with altered Purkinje cell spiking due to dysregulation of large-conductance calcium-activated potassium (BK) channel expression and function is a shared feature of disease in many degenerative ataxias. BK channel activators represent an outstanding therapeutic agent for ataxia. We have developed a high-throughput platform to screen for BK channel activators and identified a novel compound that can serve as a template for future drug development for the treatment of these disabling disorders.
Collapse
Affiliation(s)
- Sharan R Srinivasan
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Haoran Huang
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Wei-Chih Chang
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Joshua A Nasburg
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Hai M Nguyen
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Tim Strassmaier
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Heike Wulff
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| | - Vikram G Shakkottai
- Brigham and Women's Hospital, Department of Neurology, Boston, Massachusetts (S.R.S.); University of Texas Southwestern Medical Center, Department of Neurology, Dallas, Texas (H.H., V.G.S.); University of Michigan, Department of Neurology, Ann Arbor, Michigan (S.R.S., W.-C.C.); University of California, Davis, Department of Pharmacology, Davis, California (J.A.N., H.M.N., H.W.); and Nanion Technologies, Munich, Germany (T.S.)
| |
Collapse
|
30
|
Bushart DD, Shakkottai VG. Vulnerability of Human Cerebellar Neurons to Degeneration in Ataxia-Causing Channelopathies. Front Syst Neurosci 2022; 16:908569. [PMID: 35757096 PMCID: PMC9219590 DOI: 10.3389/fnsys.2022.908569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Mutations in ion channel genes underlie a number of human neurological diseases. Historically, human mutations in ion channel genes, the so-called channelopathies, have been identified to cause episodic disorders. In the last decade, however, mutations in ion channel genes have been demonstrated to result in progressive neurodegenerative and neurodevelopmental disorders in humans, particularly with ion channels that are enriched in the cerebellum. This was unexpected given prior rodent ion channel knock-out models that almost never display neurodegeneration. Human ataxia-causing channelopathies that result in even haploinsufficiency can result in cerebellar atrophy and cerebellar Purkinje neuron loss. Rodent neurons with ion channel loss-of-function appear to, therefore, be significantly more resistant to neurodegeneration compared to human neurons. Fundamental differences in susceptibility of human and rodent cerebellar neurons in ataxia-causing channelopathies must therefore be present. In this review, we explore the properties of human neurons that may contribute to their vulnerability to cerebellar degeneration secondary to ion channel loss-of-function mutations. We present a model taking into account the known allometric scaling of neuronal ion channel density in humans and other mammals that may explain the preferential vulnerability of human cerebellar neurons to degeneration in ataxia-causing channelopathies. We also speculate on the vulnerability of cerebellar neurons to degeneration in mouse models of spinocerebellar ataxia (SCA) where ion channel transcript dysregulation has recently been implicated in disease pathogenesis.
Collapse
Affiliation(s)
- David D. Bushart
- Ohio State University College of Medicine, Columbus, OH, United States
| | - Vikram G. Shakkottai
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States,*Correspondence: Vikram G. Shakkottai,
| |
Collapse
|
31
|
Mayoral-Palarz K, Neves-Carvalho A, Duarte-Silva S, Monteiro-Fernandes D, Maciel P, Khodakhah K. Cerebellar neuronal dysfunction accompanies early motor symptoms in spinocerebellar ataxia type 3. Dis Model Mech 2022; 15:275597. [PMID: 35660856 PMCID: PMC9367011 DOI: 10.1242/dmm.049514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is an adult-onset, progressive ataxia. SCA3 presents with ataxia before any gross neuropathology. A feature of many cerebellar ataxias is aberrant cerebellar output that contributes to motor dysfunction. We examined whether abnormal cerebellar output was present in the CMVMJD135 SCA3 mouse model and, if so, whether it correlated with the disease onset and progression. In vivo recordings showed that the activity of deep cerebellar nuclei neurons, the main output of the cerebellum, was altered. The aberrant activity correlated with the onset of ataxia. However, although the severity of ataxia increased with age, the severity of the aberrant cerebellar output was not progressive. The abnormal cerebellar output, however, was accompanied by non-progressive abnormal activity of their upstream synaptic inputs, the Purkinje cells. In vitro recordings indicated that alterations in intrinsic Purkinje cell pacemaking and in their synaptic inputs contributed to abnormal Purkinje cell activity. These findings implicate abnormal cerebellar physiology as an early, consistent contributor to pathophysiology in SCA3, and suggest that the aberrant cerebellar output could be an appropriate therapeutic target in SCA3. Summary: In a mouse model of spinocerebellar ataxia type 3 (SCA3), aberrant cerebellar physiology is apparent early in disease, prior to cerebellar neuronal pathology. Aberrant cerebellar output could be a therapeutic target in SCA3.
Collapse
Affiliation(s)
- Kristin Mayoral-Palarz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andreia Neves-Carvalho
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
32
|
Ransdell JL, Moreno JD, Bhagavan D, Silva JR, Nerbonne JM. Intrinsic mechanisms in the gating of resurgent Na + currents. eLife 2022; 11:e70173. [PMID: 35076394 PMCID: PMC8824471 DOI: 10.7554/elife.70173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
The resurgent component of the voltage-gated sodium current (INaR) is a depolarizing conductance, revealed on membrane hyperpolarizations following brief depolarizing voltage steps, which has been shown to contribute to regulating the firing properties of numerous neuronal cell types throughout the central and peripheral nervous systems. Although mediated by the same voltage-gated sodium (Nav) channels that underlie the transient and persistent Nav current components, the gating mechanisms that contribute to the generation of INaR remain unclear. Here, we characterized Nav currents in mouse cerebellar Purkinje neurons, and used tailored voltage-clamp protocols to define how the voltage and the duration of the initial membrane depolarization affect the amplitudes and kinetics of INaR. Using the acquired voltage-clamp data, we developed a novel Markov kinetic state model with parallel (fast and slow) inactivation pathways and, we show that this model reproduces the properties of the resurgent, as well as the transient and persistent, Nav currents recorded in (mouse) cerebellar Purkinje neurons. Based on the acquired experimental data and the simulations, we propose that resurgent Na+ influx occurs as a result of fast inactivating Nav channels transitioning into an open/conducting state on membrane hyperpolarization, and that the decay of INaR reflects the slow accumulation of recovered/opened Nav channels into a second, alternative and more slowly populated, inactivated state. Additional simulations reveal that extrinsic factors that affect the kinetics of fast or slow Nav channel inactivation and/or impact the relative distribution of Nav channels in the fast- and slow-inactivated states, such as the accessory Navβ4 channel subunit, can modulate the amplitude of INaR.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Departments of Medicine, Cardiovascular Division, Washington UniversitySt. LouisUnited States
| | - Jonathan D Moreno
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Druv Bhagavan
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Jonathan R Silva
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Jeanne M Nerbonne
- Departments of Medicine, Cardiovascular Division, Washington UniversitySt. LouisUnited States
- Developmental Biology, Washington UniversitySt. LouisUnited States
| |
Collapse
|
33
|
Hobson BD, Kong L, Angelo MF, Lieberman OJ, Mosharov EV, Herzog E, Sulzer D, Sims PA. Subcellular and regional localization of mRNA translation in midbrain dopamine neurons. Cell Rep 2022; 38:110208. [PMID: 35021090 PMCID: PMC8844886 DOI: 10.1016/j.celrep.2021.110208] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/25/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Midbrain dopaminergic (mDA) neurons exhibit extensive dendritic and axonal arborizations, but local protein synthesis is not characterized in these neurons. Here, we investigate messenger RNA (mRNA) localization and translation in mDA neuronal axons and dendrites, both of which release dopamine (DA). Using highly sensitive ribosome-bound RNA sequencing and imaging approaches, we find no evidence for mRNA translation in mDA axons. In contrast, mDA neuronal dendrites in the substantia nigra pars reticulata (SNr) contain ribosomes and mRNAs encoding the major components of DA synthesis, release, and reuptake machinery. Surprisingly, we also observe dendritic localization of mRNAs encoding synaptic vesicle-related proteins, including those involved in exocytic fusion. Our results are consistent with a role for local translation in the regulation of DA release from dendrites, but not from axons. Our translatome data define a molecular signature of sparse mDA neurons in the SNr, including the enrichment of Atp2a3/SERCA3, an atypical ER calcium pump.
Collapse
Affiliation(s)
- Benjamin D Hobson
- Department of Systems Biology, Columbia University Irving Medical Center, New York 10032, NY, USA; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Linghao Kong
- Department of Systems Biology, Columbia University Irving Medical Center, New York 10032, NY, USA
| | - Maria Florencia Angelo
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Ori J Lieberman
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Etienne Herzog
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France.
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York 10032, NY, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
34
|
Beeson KA, Westbrook GL, Schnell E. α2δ-2 is required for depolarization-induced suppression of excitation in Purkinje cells. J Physiol 2022; 600:111-122. [PMID: 34783012 PMCID: PMC8724408 DOI: 10.1113/jp282438] [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: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023] Open
Abstract
α2δ proteins (CACNA2D1-4) are required for normal neurological function and contribute to membrane trafficking of voltage-gated calcium channels, through which calcium entry initiates numerous physiological processes. However, it remains unclear how α2δ proteins influence calcium-mediated signalling to control neuronal output. Using whole-cell recordings of mouse Purkinje cells, we show that α2δ-2 is required for functional coupling of postsynaptic voltage-dependent calcium entry with calcium-dependent effector mechanisms controlling two different outputs, depolarization-induced suppression of excitation and spike afterhyperpolarization. Our findings indicate an important role for α2δ-2 proteins in regulating functional postsynaptic calcium channel coupling in neurons, providing new context for understanding the effects of α2δ mutations on neuronal circuit function and presenting additional potential avenues to manipulate α2δ-mediated signalling for therapeutic gain. KEY POINTS: Calcium influx, via voltage-dependent calcium channels, drives numerous neuronal signalling processes with precision achieved in part by tight coupling between calcium entry and calcium-dependent effectors. α2δ proteins are important for neurological function and contribute to calcium channel membrane trafficking, although how α2δ proteins influence postsynaptic calcium-dependent signalling is largely unexplored. Here it is shown that loss of α2δ-2 proteins disrupts functional calcium coupling to two different postsynaptic calcium-dependent signals in mouse Purkinje cell neurons, retrograde endocannabinoid signalling and the action potential afterhyperpolarization. The findings provide new insights into the control of calcium coupling as well as new roles for α2δ-2 proteins in neurons.
Collapse
Affiliation(s)
- Kathleen A. Beeson
- Neuroscience Graduate Program, OHSU, Portland, OR, 97239,Department of Anesthesiology and Perioperative Medicine, OHSU, Portland, OR, 97239
| | | | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, OHSU, Portland, OR, 97239,Operative Care Division, Portland VA Health Care System, Portland, OR, 97239,Eric Schnell, MD, PhD,
| |
Collapse
|
35
|
González-Tapia D, Vázquez-Hernández N, Urmeneta-Ortiz F, Navidad-Hernandez N, Lazo-Yepez M, Tejeda-Martínez A, Flores-Soto M, González-Burgos I. 3-Acetylpyridine-induced ataxic-like motor impairments are associated with plastic changes in the Purkinje cells of the rat cerebellum. Neurologia 2021. [DOI: 10.1016/j.nrl.2021.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
36
|
Poelzing S, Weinberg SH, Keener JP. Initiation and entrainment of multicellular automaticity via diffusion limited extracellular domains. Biophys J 2021; 120:5279-5294. [PMID: 34757078 DOI: 10.1016/j.bpj.2021.10.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 09/12/2021] [Accepted: 10/26/2021] [Indexed: 01/07/2023] Open
Abstract
Electrically excitable cells often spontaneously and synchronously depolarize in vitro and in vivo preparations. It remains unclear how cells entrain and autorhythmically activate above the intrinsic mean activation frequency of isolated cells with or without pacemaking mechanisms. Recent studies suggest that cyclic ion accumulation and depletion in diffusion-limited extracellular volumes modulate electrophysiology by ephaptic mechanisms (nongap junction or synaptic coupling). This report explores how potassium accumulation and depletion in a restricted extracellular domain induces spontaneous action potentials in two different computational models of excitable cells without gap junctional coupling: Hodgkin-Huxley and Luo-Rudy. Importantly, neither model will spontaneously activate on its own without external stimuli. Simulations demonstrate that cells sharing a diffusion-limited extracellular compartment can become autorhythmic and entrained despite intercellular electrical heterogeneity. Autorhythmic frequency is modulated by the cleft volume and potassium fluxes through the cleft. Additionally, inexcitable cells can suppress or induce autorhythmic activity in an excitable cell via a shared cleft. Diffusion-limited shared clefts can also entrain repolarization. Critically, this model predicts a mechanism by which diffusion-limited shared clefts can initiate, entrain, and modulate multicellular automaticity in the absence of gap junctions.
Collapse
Affiliation(s)
- Steven Poelzing
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Heart and Reparative Medicine, and the Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Roanoke, Virginia.
| | - Seth H Weinberg
- Department of Biomedical Engineering, Davis Heart and Lung Research Institute, and the Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - James P Keener
- Department of Mathematics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
37
|
van der Heijden ME, Lackey EP, Perez R, Ișleyen FS, Brown AM, Donofrio SG, Lin T, Zoghbi HY, Sillitoe RV. Maturation of Purkinje cell firing properties relies on neurogenesis of excitatory neurons. eLife 2021; 10:e68045. [PMID: 34542409 PMCID: PMC8452305 DOI: 10.7554/elife.68045] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/31/2021] [Indexed: 01/18/2023] Open
Abstract
Preterm infants that suffer cerebellar insults often develop motor disorders and cognitive difficulty. Excitatory granule cells, the most numerous neuron type in the brain, are especially vulnerable and likely instigate disease by impairing the function of their targets, the Purkinje cells. Here, we use regional genetic manipulations and in vivo electrophysiology to test whether excitatory neurons establish the firing properties of Purkinje cells during postnatal mouse development. We generated mutant mice that lack the majority of excitatory cerebellar neurons and tracked the structural and functional consequences on Purkinje cells. We reveal that Purkinje cells fail to acquire their typical morphology and connectivity, and that the concomitant transformation of Purkinje cell firing activity does not occur either. We also show that our mutant pups have impaired motor behaviors and vocal skills. These data argue that excitatory cerebellar neurons define the maturation time-window for postnatal Purkinje cell functions and refine cerebellar-dependent behaviors.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Elizabeth P Lackey
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Ross Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Fatma S Ișleyen
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| | - Amanda M Brown
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Sarah G Donofrio
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Tao Lin
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
38
|
Markanday A, Inoue J, Dicke PW, Thier P. Cerebellar complex spikes multiplex complementary behavioral information. PLoS Biol 2021; 19:e3001400. [PMID: 34529650 PMCID: PMC8478165 DOI: 10.1371/journal.pbio.3001400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 09/28/2021] [Accepted: 08/25/2021] [Indexed: 12/02/2022] Open
Abstract
Purkinje cell (PC) discharge, the only output of cerebellar cortex, involves 2 types of action potentials, high-frequency simple spikes (SSs) and low-frequency complex spikes (CSs). While there is consensus that SSs convey information needed to optimize movement kinematics, the function of CSs, determined by the PC’s climbing fiber input, remains controversial. While initially thought to be specialized in reporting information on motor error for the subsequent amendment of behavior, CSs seem to contribute to other aspects of motor behavior as well. When faced with the bewildering diversity of findings and views unraveled by highly specific tasks, one may wonder if there is just one true function with all the other attributions wrong? Or is the diversity of findings a reflection of distinct pools of PCs, each processing specific streams of information conveyed by climbing fibers? With these questions in mind, we recorded CSs from the monkey oculomotor vermis deploying a repetitive saccade task that entailed sizable motor errors as well as small amplitude saccades, correcting them. We demonstrate that, in addition to carrying error-related information, CSs carry information on the metrics of both primary and small corrective saccades in a time-specific manner, with changes in CS firing probability coupled with changes in CS duration. Furthermore, we also found CS activity that seemed to predict the upcoming events. Hence PCs receive a multiplexed climbing fiber input that merges complementary streams of information on the behavior, separable by the recipient PC because they are staggered in time. Purkinje cell (PC) discharge, the only output of cerebellar cortex, involves both high-frequency simple spikes and low-frequency complex spikes; the function of the latter, determined by a PC’s climbing fibre input, remains controversial. This study shows that PCs receive a multiplexed climbing fibre input that merges complementary streams of information relevant for behaviour.
Collapse
Affiliation(s)
- Akshay Markanday
- Hertie Institute for Clinical Brain Research, Tübingen, Germany
- Graduate School of Neural and Behavioral Sciences, International Max Planck Research School, Tübingen University, Tübingen, Germany
- * E-mail: (AM); (PT)
| | - Junya Inoue
- Graduate School of Neural and Behavioral Sciences, International Max Planck Research School, Tübingen University, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen University, Tübingen, Germany
| | - Peter W. Dicke
- Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Peter Thier
- Hertie Institute for Clinical Brain Research, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen University, Tübingen, Germany
- * E-mail: (AM); (PT)
| |
Collapse
|
39
|
Brandenburg C, Smith LA, Kilander MBC, Bridi MS, Lin YC, Huang S, Blatt GJ. Parvalbumin subtypes of cerebellar Purkinje cells contribute to differential intrinsic firing properties. Mol Cell Neurosci 2021; 115:103650. [PMID: 34197921 DOI: 10.1016/j.mcn.2021.103650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 01/26/2023] Open
Abstract
Purkinje cells (PCs) are central to cerebellar information coding and appreciation for the diversity of their firing patterns and molecular profiles is growing. Heterogeneous subpopulations of PCs have been identified that display differences in intrinsic firing properties without clear mechanistic insight into what underlies the divergence in firing parameters. Although long used as a general PC marker, we report that the calcium binding protein parvalbumin labels a subpopulation of PCs, based on high and low expression, with a conserved distribution pattern across the animals examined. We trained a convolutional neural network to recognize the parvalbumin subtypes and create maps of whole cerebellar distribution and find that PCs within these areas have differences in spontaneous firing that can be modified by altering calcium buffer content. These subtypes also show differential responses to potassium and calcium channel blockade, suggesting a mechanistic role for variability in PC intrinsic firing through differences in ion channel composition. It is proposed that ion channels drive the diversity in PC intrinsic firing phenotype and parvalbumin calcium buffering provides capacity for the highest firing rates observed. These findings open new avenues for detailed classification of PC subtypes.
Collapse
Affiliation(s)
- Cheryl Brandenburg
- Hussman Institute for Autism, Baltimore, MD 21201, USA; University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | - Yu-Chih Lin
- Hussman Institute for Autism, Baltimore, MD 21201, USA
| | - Shiyong Huang
- Hussman Institute for Autism, Baltimore, MD 21201, USA.
| | - Gene J Blatt
- Hussman Institute for Autism, Baltimore, MD 21201, USA.
| |
Collapse
|
40
|
Beekhof GC, Osório C, White JJ, van Zoomeren S, van der Stok H, Xiong B, Nettersheim IH, Mak WA, Runge M, Fiocchi FR, Boele HJ, Hoebeek FE, Schonewille M. Differential spatiotemporal development of Purkinje cell populations and cerebellum-dependent sensorimotor behaviors. eLife 2021; 10:63668. [PMID: 33973524 PMCID: PMC8195607 DOI: 10.7554/elife.63668] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
Distinct populations of Purkinje cells (PCs) with unique molecular and connectivity features are at the core of the modular organization of the cerebellum. Previously, we showed that firing activity of PCs differs between ZebrinII-positive and ZebrinII-negative cerebellar modules (Zhou et al., 2014; Wu et al., 2019). Here, we investigate the timing and extent of PC differentiation during development in mice. We found that several features of PCs, including activity levels, dendritic arborization, axonal shape and climbing fiber input, develop differentially between nodular and anterior PC populations. Although all PCs show a particularly rapid development in the second postnatal week, anterior PCs typically have a prolonged physiological and dendritic maturation. In line herewith, younger mice exhibit attenuated anterior-dependent eyeblink conditioning, but faster nodular-dependent compensatory eye movement adaptation. Our results indicate that specific cerebellar regions have unique developmental timelines which match with their related, specific forms of cerebellum-dependent behaviors.
Collapse
Affiliation(s)
| | - Catarina Osório
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Joshua J White
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Bilian Xiong
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Marit Runge
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Henk-Jan Boele
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Princeton Neuroscience Institute, Princeton, United States
| | - Freek E Hoebeek
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht, Netherlands
| | | |
Collapse
|
41
|
Chan WK, Fetit R, Griffiths R, Marshall H, Mason JO, Price DJ. Using organoids to study human brain development and evolution. Dev Neurobiol 2021; 81:608-622. [PMID: 33773072 DOI: 10.1002/dneu.22819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/22/2022]
Abstract
Recent advances in methods for making cerebral organoids have opened a window of opportunity to directly study human brain development and disease, countering limitations inherent in non-human-based approaches. Whether freely patterned, guided into a region-specific fate or fused into assembloids, organoids have successfully recapitulated key features of in vivo neurodevelopment, allowing its examination from early to late stages. Although organoids have enormous potential, their effective use relies on understanding the extent of their limitations in accurately reproducing specific processes and components in the developing human brain. Here we review the potential of cerebral organoids to model and study human brain development and evolution and discuss the progress and current challenges in their use for reproducing specific human neurodevelopmental processes.
Collapse
Affiliation(s)
- Wai-Kit Chan
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Rana Fetit
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Rosie Griffiths
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Helen Marshall
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - John O Mason
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - David J Price
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
42
|
Nilsson MNP, Jörntell H. Channel current fluctuations conclusively explain neuronal encoding of internal potential into spike trains. Phys Rev E 2021; 103:022407. [PMID: 33736029 DOI: 10.1103/physreve.103.022407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/15/2021] [Indexed: 11/07/2022]
Abstract
Hodgkin and Huxley's seminal neuron model describes the propagation of voltage spikes in axons, but it cannot explain certain full-neuron features crucial for understanding the neural code. We consider channel current fluctuations in a trisection of the Hodgkin-Huxley model, allowing an analytic-mechanistic explanation of these features and yielding consistently excellent matches with in vivo recordings of cerebellar Purkinje neurons, which we use as model systems. This shows that the neuronal encoding is described conclusively by a soft-thresholding function having just three parameters.
Collapse
Affiliation(s)
| | - Henrik Jörntell
- Department of Experimental Medical Science, Lund University, SE-221 00 Lund, Sweden
| |
Collapse
|
43
|
Gating by Functionally Indivisible Cerebellar Circuits: a Hypothesis. THE CEREBELLUM 2021; 20:518-532. [PMID: 33464470 PMCID: PMC8360902 DOI: 10.1007/s12311-020-01223-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 11/08/2022]
Abstract
The attempt to understand the cerebellum has been dominated for years by supervised learning models. The central idea is that a learning algorithm modifies transmission strength at repeatedly co-active synapses, creating memories stored as finely calibrated synaptic weights. As a result, Purkinje cells, usually the de facto output cells of these models, acquire a modified response to input in a remembered pattern. This paper proposes an alternative model of pattern memory in which the function of a match is permissive, allowing but not driving output, and accordingly controlling the timing of output but not the rate of firing by Purkinje cells. Learning does not result in graded synaptic weights. There is no supervised learning algorithm or memory of individual patterns, which, like graded weights, are unnecessary to explain the evidence. Instead, patterns are classed as simply either known or not, at the level of input to a functional population of 100s of Purkinje cells (a microzone). The standard is strict. If only a handful of Purkinje cells receive a mismatch output of the whole circuit is blocked. Only if there is a full and accurate match are projection neurons in deep nuclei, which carry the output of most circuits, released from default inhibitory restraint. Purkinje cell firing at those times is a linear function of input rates. There is no effect of modification of synaptic transmission except to either allow or block output.
Collapse
|
44
|
Liu Y, Xing H, Wilkes BJ, Yokoi F, Chen H, Vaillancourt DE, Li Y. The abnormal firing of Purkinje cells in the knockin mouse model of DYT1 dystonia. Brain Res Bull 2020; 165:14-22. [PMID: 32976982 PMCID: PMC7674218 DOI: 10.1016/j.brainresbull.2020.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/23/2020] [Accepted: 09/13/2020] [Indexed: 12/27/2022]
Abstract
DYT1 dystonia is an inherited movement disorder caused by a heterozygous trinucleotide (GAG) deletion in DYT1/TOR1A, coding for torsinA. Growing evidence suggests that the cerebellum plays a role in the pathogenesis of dystonia. Brain imaging of both DYT1 dystonia patients and animal models show abnormal activity in the cerebellum. The cerebellum-specific knockdown of torsinA in adult mice leads to dystonia-like behavior. Dyt1 ΔGAG heterozygous knock-in mouse model exhibits impaired corticostriatal long-term depression, abnormal muscle co-contraction, and motor deficits. We and others previously reported altered dendritic structures in Purkinje cells in Dyt1 knock-in mouse models. However, whether there are any electrophysiological alterations of the Purkinje cells in Dyt1 knock-in mice is not known. We used the patch-clamp recording in brain slices and in acutely dissociated Purkinje cells to identify specific alterations of Purkinje cells firing. We found abnormal firing of non-tonic type of Purkinje cells in the Dyt1 knock-in mice. Furthermore, the large-conductance calcium-activated potassium (BK) current and the BK channel protein levels were significantly increased in the Dyt1 knock-in mice. Our results support a role of the cerebellum in the pathogenesis of DYT1 dystonia. Manipulating the Purkinje cell firing and cerebellar output may show great promise for treating DYT1 dystonia.
Collapse
Affiliation(s)
- Yuning Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA; Genetics Institute, University of Florida, University of Florida, Gainesville, FL, USA
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Bradley J Wilkes
- Department of Applied Physiology and Kinesiology, Biomedical Engineering, and Neurology, University of Florida, Gainesville, FL, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Huanxin Chen
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, Biomedical Engineering, and Neurology, University of Florida, Gainesville, FL, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
45
|
Chopra R, Bushart DD, Cooper JP, Yellajoshyula D, Morrison LM, Huang H, Handler HP, Man LJ, Dansithong W, Scoles DR, Pulst SM, Orr HT, Shakkottai VG. Altered Capicua expression drives regional Purkinje neuron vulnerability through ion channel gene dysregulation in spinocerebellar ataxia type 1. Hum Mol Genet 2020; 29:3249-3265. [PMID: 32964235 PMCID: PMC7689299 DOI: 10.1093/hmg/ddaa212] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/19/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
Selective neuronal vulnerability in neurodegenerative disease is poorly understood. Using the ATXN1[82Q] model of spinocerebellar ataxia type 1 (SCA1), we explored the hypothesis that regional differences in Purkinje neuron degeneration could provide novel insights into selective vulnerability. ATXN1[82Q] Purkinje neurons from the anterior cerebellum were found to degenerate earlier than those from the nodular zone, and this early degeneration was associated with selective dysregulation of ion channel transcripts and altered Purkinje neuron spiking. Efforts to understand the basis for selective dysregulation of channel transcripts revealed modestly increased expression of the ATXN1 co-repressor Capicua (Cic) in anterior cerebellar Purkinje neurons. Importantly, disrupting the association between ATXN1 and Cic rescued the levels of these ion channel transcripts, and lentiviral overexpression of Cic in the nodular zone accelerated both aberrant Purkinje neuron spiking and neurodegeneration. These findings reinforce the central role for Cic in SCA1 cerebellar pathophysiology and suggest that only modest reductions in Cic are needed to have profound therapeutic impact in SCA1.
Collapse
Affiliation(s)
- Ravi Chopra
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Neurology, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - David D Bushart
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - John P Cooper
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Logan M Morrison
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Haoran Huang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hillary P Handler
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Luke J Man
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Warunee Dansithong
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Daniel R Scoles
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
46
|
Cerebral Cortical Activity Following Non-invasive Cerebellar Stimulation-a Systematic Review of Combined TMS and EEG Studies. THE CEREBELLUM 2020; 19:309-335. [PMID: 31907864 DOI: 10.1007/s12311-019-01093-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The cerebellum sends dense projections to both motor and non-motor regions of the cerebral cortex via the cerebellarthalamocortical tract. The integrity of this tract is crucial for healthy motor and cognitive function. This systematic review examines research using transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS) to the cerebellum with combined cortical electroencephalography (EEG) to explore the temporal features of cerebellar-cortical connectivity. A detailed discussion of the outcomes and limitations of the studies meeting review criteria is presented. Databases were searched between 1 December 2017 and 6 December 2017, with Scopus alerts current as of 23 July 2019. Of the 407 studies initially identified, 10 met review criteria. Findings suggested that cerebellar-cortical assessment is suited to combined TMS and EEG, although work is required to ensure experimental procedures are optimal for eliciting a reliable cerebellar response from stimulation. A distinct variation in methodologies and outcome measures employed across studies, and small sample sizes limited the conclusions that could be drawn regarding the electrophysiological signatures of cerebellar-cortical communication. This review highlights the need for stringent protocols and methodologies for cerebellar-cortical assessments via combined TMS and EEG. With these in place, combined TMS and EEG will provide a valuable means for exploring cerebellar connectivity with a wide range of cortical sites. Assessments have the potential to aid in the understanding of motor and cognitive function in both healthy and clinical groups, and provide insights into long-range neural communication generally.
Collapse
|
47
|
In vivo analysis of the spontaneous firing of cerebellar Purkinje cells in awake transgenic mice that model spinocerebellar ataxia type 2. Cell Calcium 2020; 93:102319. [PMID: 33248384 DOI: 10.1016/j.ceca.2020.102319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022]
Abstract
Cerebellar Purkinje cells (PCs) fire spontaneously in a tonic mode, although the precision of this pacemaking activity is disturbed in many abnormal conditions involving cerebellar atrophy, such as many spinocerebellar ataxias (SCAs). In our previous studies we used the single-unit extracellular recording method to analyze spontaneous PC firing in vivo in the anesthetized SCA2-58Q transgenic mice. We realized that PCs from aging SCA2-58Q mice fire much less regularly compared to PCs from their wild type (WT) littermates and this abnormal activity can be reversed with an intraperitoneal (i. p.) injection of SK channel-positive modulator chlorzoxazone (CHZ). Here we used the same single-unit extracellular recording method to analyze the spontaneous firing in vivo in awake SCA2-58Q transgenic mice. For this purpose, we used the Mobile HomeCage (Neurotar, Finland) floating platform to immobilize the experimental animal's head during the recording sessions. We discovered that generally PCs from awake animals fired much more frequently and much less regularly than previously observed PCs from anesthetized animals. In vivo recordings from awake SCA2/WT mice revealed that complex spikes, which are generated by PCs in reply to the excitation coming by climbing fibers, as well as simple spikes, were much less frequent in SCA2 mice compared to their WT littermates. To test the effect of the SK channel positive modulation on the PCs firing activity in awake SCA2 mice and also the effect on their motor coordination, we started the CHZ trial in these mice. We discovered that the long-term i. p. injections of CHZ did not affect the spike generation in SCA2-58Q mice, however, they did recover the precision of this spontaneous pacemaking activity. Furthermore, we also showed that treatment with CHZ alleviated the age-dependent motor impairment in SCA2-58Q mice. We propose that the lack of precision in PC spike generation might be a key cause for the progression of ataxic symptoms in different SCAs and that the activation of calcium-activated potassium channels, including SK channels, can be used as a potential way to treat SCAs on the physiological level of the disease.
Collapse
|
48
|
Neves-Carvalho A, Duarte-Silva S, Teixeira-Castro A, Maciel P. Polyglutamine spinocerebellar ataxias: emerging therapeutic targets. Expert Opin Ther Targets 2020; 24:1099-1119. [PMID: 32962458 DOI: 10.1080/14728222.2020.1827394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Six of the most frequent dominantly inherited spinocerebellar ataxias (SCAs) worldwide - SCA1, SCA2, SCA3, SCA6, SCA7, and SCA17 - are caused by an expansion of a polyglutamine (polyQ) tract in the corresponding proteins. While the identification of the causative mutation has advanced knowledge on the pathogenesis of polyQ SCAs, effective therapeutics able to mitigate the severe clinical manifestation of these highly incapacitating disorders are not yet available. AREAS COVERED This review provides a comprehensive and critical perspective on well-established and emerging therapeutic targets for polyQ SCAs; it aims to inspire prospective drug discovery efforts. EXPERT OPINION The landscape of polyQ SCAs therapeutic targets and strategies includes (1) the mutant genes and proteins themselves, (2) enhancement of endogenous protein quality control responses, (3) abnormal protein-protein interactions of the mutant proteins, (4) disturbed neuronal function, (5) mitochondrial function, energy availability and oxidative stress, and (6) glial dysfunction, growth factor or hormone imbalances. Challenges include gaining a clearer definition of therapeutic targets for the drugs in clinical development, the discovery of novel drug-like molecules for challenging key targets, and the attainment of a stronger translation of preclinical findings to the clinic.
Collapse
Affiliation(s)
- Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| |
Collapse
|
49
|
Yamamoto M, Kim M, Imai H, Itakura Y, Ohtsuki G. Microglia-Triggered Plasticity of Intrinsic Excitability Modulates Psychomotor Behaviors in Acute Cerebellar Inflammation. Cell Rep 2020; 28:2923-2938.e8. [PMID: 31509752 DOI: 10.1016/j.celrep.2019.07.078] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/20/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cerebellar dysfunction relates to various psychiatric disorders, including autism spectrum and depressive disorders. However, the physiological aspect is less advanced. Here, we investigate the immune-triggered hyperexcitability in the cerebellum on a wider scope. Activated microglia via exposure to bacterial endotoxin lipopolysaccharide or heat-killed Gram-negative bacteria induce a potentiation of the intrinsic excitability in Purkinje neurons, which is suppressed by microglia-activity inhibitor and microglia depletion. An inflammatory cytokine, tumor necrosis factor alpha (TNF-α), released from microglia via toll-like receptor 4, triggers this plasticity. Our two-photon FRET ATP imaging shows an increase in ATP concentration following endotoxin exposure. Both TNF-α and ATP secretion facilitate synaptic transmission. Region-specific inflammation in the cerebellum in vivo shows depression- and autistic-like behaviors. Furthermore, both TNF-α inhibition and microglia depletion revert such behavioral abnormality. Resting-state functional MRI reveals overconnectivity between the inflamed cerebellum and the prefrontal neocortical regions. Thus, immune activity in the cerebellum induces neuronal hyperexcitability and disruption of psychomotor behaviors in animals.
Collapse
Affiliation(s)
- Masamichi Yamamoto
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto University Hospital, Shogoin-Kawaramachi-cho, Sakyo-ward, Kyoto 606-8507, Japan
| | - Minsoo Kim
- The Hakubi Center for Advanced Research, Kyoto University, Yoshida, Sakyo-ward, Kyoto 606-8501, Japan; Department of Molecular and Cellular Physiology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ward, Kyoto 606-8501, Japan
| | - Hirohiko Imai
- Department of Systems Science, Kyoto University Graduate School of Informatics, Yoshida-Honmachi, Sakyo-ward, Kyoto 606-8501, Japan
| | - Yamato Itakura
- Department of Biophysics, Kyoto University Graduate School of Science, Kitashirakawa-Oiwake-cho, Sakyo-ward, Kyoto 606-8502, Japan
| | - Gen Ohtsuki
- The Hakubi Center for Advanced Research, Kyoto University, Yoshida, Sakyo-ward, Kyoto 606-8501, Japan; Department of Biophysics, Kyoto University Graduate School of Science, Kitashirakawa-Oiwake-cho, Sakyo-ward, Kyoto 606-8502, Japan.
| |
Collapse
|
50
|
Uddin LQ. Bring the Noise: Reconceptualizing Spontaneous Neural Activity. Trends Cogn Sci 2020; 24:734-746. [PMID: 32600967 PMCID: PMC7429348 DOI: 10.1016/j.tics.2020.06.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
Definitions of what constitutes the 'signal of interest' in neuroscience can be controversial, due in part to continuously evolving notions regarding the significance of spontaneous neural activity. This review highlights how the challenge of separating brain signal from noise has led to new conceptualizations of brain functional organization at both the micro- and macroscopic level. Recent debates in the functional neuroimaging community surrounding artifact removal processes have revived earlier discussions surrounding how to appropriately isolate and measure neuronal signals against a background of noise from various sources. Insights from electrophysiological studies and computational modeling can inform current theory and data analytic practices in human functional neuroimaging, given that signal and noise may be inextricably linked in the brain.
Collapse
Affiliation(s)
- Lucina Q Uddin
- Department of Psychology, University of Miami, PO Box 248185-0751, Coral Gables, FL 33124, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|