1
|
Brown SP, Jena AK, Osko JJ, Ransdell JL. Tsc1 Deletion in Purkinje Neurons Disrupts the Axon Initial Segment, Impairing Excitability and Cerebellar Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635932. [PMID: 39974887 PMCID: PMC11838410 DOI: 10.1101/2025.01.31.635932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Loss-of-function mutations in tuberous sclerosis 1 ( TSC1 ) are prevalent monogenic causes of autism spectrum disorder (ASD). Selective deletion of Tsc1 from mouse cerebellar Purkinje neurons has been shown to cause several ASD-linked behavioral impairments, which are linked to reduced Purkinje neuron repetitive firing rates. We used electrophysiology methods to investigate why Purkinje neuron-specific Tsc1 deletion ( Tsc1 mut/mut ) impairs Purkinje neuron firing. These studies revealed a depolarized shift in action potential threshold voltage, an effect that we link to reduced expression of the fast-transient voltage-gated sodium (Nav) current in Tsc1 mut/mut Purkinje neurons. The reduced Nav currents in these cells was associated with diminished secondary immunofluorescence from anti-pan Nav channel labeling at Purkinje neuron axon initial segments (AIS). Interestingly, anti-ankyrinG immunofluorescence was also found to be significantly reduced at the AIS of Tsc1 mut/mut Purkinje neurons, suggesting Tsc1 is necessary for the organization and functioning of the Purkinje neuron AIS. An analysis of the 1 st and 2 nd derivative of the action potential voltage-waveform supported this hypothesis, revealing spike initiation and propagation from the AIS of Tsc1 mut/mut Purkinje neurons is impaired compared to age-matched control Purkinje neurons. Heterozygous Tsc1 deletion resulted in no significant changes in the firing properties of adult Purkinje neurons, and slight reductions in anti-pan Nav and anti-ankyrinG labeling at the Purkinje neuron AIS, revealing deficits in Purkinje neuron firing due to Tsc1 haploinsufficiency are delayed compared to age-matched Tsc1 mut/mut Purkinje neurons. Together, these data reveal the loss of Tsc1 impairs Purkinje neuron firing and membrane excitability through the dysregulation of proteins necessary for AIS organization and function.
Collapse
|
2
|
Espino CM, Nagaraja C, Ortiz S, Dayton JR, Murali AR, Ma Y, Mann EL, Garlapalli S, Wohlgemuth RP, Brashear SE, Smith LR, Wilkinson KA, Griffith TN. Differential encoding of mammalian proprioception by voltage-gated sodium channels. SCIENCE ADVANCES 2025; 11:eads6660. [PMID: 39772670 PMCID: PMC11708877 DOI: 10.1126/sciadv.ads6660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Animals requiring purposeful movement for survival are endowed with mechanoreceptors, called proprioceptors, that provide essential sensory feedback from muscles and joints to spinal cord circuits, which modulates motor output. Despite the essential nature of proprioceptive signaling in daily life, the mechanisms governing proprioceptor activity are poorly understood. Here, we identified nonredundant roles for two voltage-gated sodium channels (NaVs), NaV1.1 and NaV1.6, in mammalian proprioception. Deletion of NaV1.6 in somatosensory neurons (NaV1.6cKO mice) causes severe motor deficits accompanied by loss of proprioceptive transmission, which contrasts with our previous findings using similar mouse models to target NaV1.1 (NaV1.1cKO). In NaV1.6cKO animals, we observed impairments in proprioceptor end-organ structure and a marked reduction in skeletal muscle myofiber size that were absent in NaV1.1cKO mice. We attribute the differential contributions of NaV1.1 and NaV1.6 to distinct cellular localization patterns. Collectively, we provide evidence that NaVs uniquely shape neural signaling within a somatosensory modality.
Collapse
Affiliation(s)
- Cyrrus M. Espino
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Chetan Nagaraja
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Serena Ortiz
- Department of Biological Sciences, San José State University, San Jose, CA, USA
| | - Jacquelyn R. Dayton
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Akash R. Murali
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Yanki Ma
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Emari L. Mann
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Postbaccalaureate Research Education Program at UC Davis, University of California, Davis, Davis, CA, USA
| | - Snigdha Garlapalli
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Psychology, University of California, Davis, Davis, CA, USA
| | - Ross P. Wohlgemuth
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Sarah E. Brashear
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Lucas R. Smith
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | | | - Theanne N. Griffith
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| |
Collapse
|
3
|
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
|
4
|
Johnson JP, Focken T, Karimi Tari P, Dube C, Goodchild SJ, Andrez JC, Bankar G, Burford K, Chang E, Chowdhury S, Christabel J, Dean R, de Boer G, Dehnhardt C, Gong W, Grimwood M, Hussainkhel A, Jia Q, Khakh K, Lee S, Li J, Lin S, Lindgren A, Lofstrand V, Mezeyova J, Nelkenbrecher K, Shuart NG, Sojo L, Sun S, Waldbrook M, Wesolowski S, Wilson M, Xie Z, Zenova A, Zhang W, Scott FL, Cutts AJ, Sherrington RP, Winquist R, Cohen CJ, Empfield JR. The contribution of Na V1.6 to the efficacy of voltage-gated sodium channel inhibitors in wild type and Na V1.6 gain-of-function (GOF) mouse seizure control. Br J Pharmacol 2024; 181:3993-4011. [PMID: 38922847 DOI: 10.1111/bph.16481] [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: 04/19/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND AND PURPOSE Inhibitors of voltage-gated sodium channels (NaVs) are important anti-epileptic drugs, but the contribution of specific channel isoforms is unknown since available inhibitors are non-selective. We aimed to create novel, isoform selective inhibitors of Nav channels as a means of informing the development of improved antiseizure drugs. EXPERIMENTAL APPROACH We created a series of compounds with diverse selectivity profiles enabling block of NaV1.6 alone or together with NaV1.2. These novel NaV inhibitors were evaluated for their ability to inhibit electrically evoked seizures in mice with a heterozygous gain-of-function mutation (N1768D/+) in Scn8a (encoding NaV1.6) and in wild-type mice. KEY RESULTS Pharmacologic inhibition of NaV1.6 in Scn8aN1768D/+ mice prevented seizures evoked by a 6-Hz shock. Inhibitors were also effective in a direct current maximal electroshock seizure assay in wild-type mice. NaV1.6 inhibition correlated with efficacy in both models, even without inhibition of other CNS NaV isoforms. CONCLUSIONS AND IMPLICATIONS Our data suggest NaV1.6 inhibition is a driver of efficacy for NaV inhibitor anti-seizure medicines. Sparing the NaV1.1 channels of inhibitory interneurons did not compromise efficacy. Selective NaV1.6 inhibitors may provide targeted therapies for human Scn8a developmental and epileptic encephalopathies and improved treatments for idiopathic epilepsies.
Collapse
Affiliation(s)
- James P Johnson
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Thilo Focken
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Parisa Karimi Tari
- Department of In Vivo Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Celine Dube
- Department of In Vivo Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Samuel J Goodchild
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | | | - Girish Bankar
- Department of In Vivo Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Kristen Burford
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Elaine Chang
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Sultan Chowdhury
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Jessica Christabel
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Richard Dean
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Gina de Boer
- Department of Compound Properties, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Christoph Dehnhardt
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Wei Gong
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Michael Grimwood
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Angela Hussainkhel
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Qi Jia
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Kuldip Khakh
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Stephanie Lee
- Department of Compound Properties, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Jenny Li
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Sophia Lin
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Andrea Lindgren
- Department of Compound Properties, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Verner Lofstrand
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Janette Mezeyova
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Karen Nelkenbrecher
- Department of In Vivo Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Noah Gregory Shuart
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Luis Sojo
- Department of Compound Properties, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Shaoyi Sun
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Matthew Waldbrook
- Department of In Vivo Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Steven Wesolowski
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Michael Wilson
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Zhiwei Xie
- Department of In Vitro Biology, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Alla Zenova
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Wei Zhang
- Department of Chemistry, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | | | - Alison J Cutts
- Scientific Affairs, Xenon Pharmaceuticals, Inc, Burnaby, British Columbia, Canada
| | - Robin P Sherrington
- Executive Team, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Raymond Winquist
- Executive Team, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - Charles J Cohen
- Executive Team, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| | - James R Empfield
- Executive Team, Xenon Pharmaceuticals Inc, Burnaby, British Columbia, Canada
| |
Collapse
|
5
|
Yu W, Hill SF, Zhu L, Demetriou Y, Reger F, Mattis J, Meisler MH. Dentate gyrus granule cells are a locus of pathology in Scn8a developmental encephalopathy. Neurobiol Dis 2024; 199:106591. [PMID: 38969233 DOI: 10.1016/j.nbd.2024.106591] [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: 01/26/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024] Open
Abstract
Gain-of-function mutations in SCN8A cause developmental and epileptic encephalopathy (DEE), a disorder characterized by early-onset refractory seizures, deficits in motor and intellectual functions, and increased risk of sudden unexpected death in epilepsy. Altered activity of neurons in the corticohippocampal circuit has been reported in mouse models of DEE. We examined the effect of chronic seizures on gene expression in the hippocampus by single-nucleus RNA sequencing in mice expressing the patient mutation SCN8A-p.Asn1768Asp (N1768D). One hundred and eighty four differentially expressed genes were identified in dentate gyrus granule cells, many more than in other cell types. Electrophysiological recording from dentate gyrus granule cells demonstrated an elevated firing rate. Targeted reduction of Scn8a expression in the dentate gyrus by viral delivery of an shRNA resulted in doubling of median survival time from 4 months to 8 months, whereas delivery of shRNA to the CA1 and CA3 regions did not result in lengthened survival. These data indicate that granule cells of the dentate gyrus are a specific locus of pathology in SCN8A-DEE.
Collapse
Affiliation(s)
- Wenxi Yu
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Sophie F Hill
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Limei Zhu
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | - Faith Reger
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Joanna Mattis
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Espino CM, Nagaraja C, Ortiz S, Dayton JR, Murali AR, Ma Y, Mann EL, Garlapalli S, Wohlgemuth RP, Brashear SE, Smith LR, Wilkinson KA, Griffith TN. Differential encoding of mammalian proprioception by voltage-gated sodium channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609982. [PMID: 39253497 PMCID: PMC11383322 DOI: 10.1101/2024.08.27.609982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Animals that require purposeful movement for survival are endowed with mechanosensory neurons called proprioceptors that provide essential sensory feedback from muscles and joints to spinal cord circuits, which modulates motor output. Despite the essential nature of proprioceptive signaling in daily life, the mechanisms governing proprioceptor activity are poorly understood. Here, we have identified distinct and nonredundant roles for two voltage-gated sodium channels (NaVs), NaV1.1 and NaV1.6, in mammalian proprioception. Deletion of NaV1.6 in somatosensory neurons (NaV1.6cKO mice) causes severe motor deficits accompanied by complete loss of proprioceptive transmission, which contrasts with our previous findings using similar mouse models to target NaV1.1 (NaV1.1cKO). In NaV1.6cKO animals, loss of proprioceptive feedback caused non-cell-autonomous impairments in proprioceptor end-organs and skeletal muscle that were absent in NaV1.1cKO mice. We attribute the differential contribution of NaV1.1 and NaV1.6 in proprioceptor function to distinct cellular localization patterns. Collectively, these data provide the first evidence that NaV subtypes uniquely shape neurotransmission within a somatosensory modality.
Collapse
Affiliation(s)
- Cyrrus M. Espino
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Chetan Nagaraja
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Serena Ortiz
- Department of Biological Sciences, San José State University, San Jose, CA, USA
| | - Jacquelyn R. Dayton
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| | - Akash R. Murali
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Yanki Ma
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Emari L. Mann
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Postbaccalaureate Research Education Program at UC Davis, University of California, Davis, Davis, CA, USA
| | - Snigdha Garlapalli
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
- Undergraduate Program in Psychology, University of California, Davis, Davis, CA, USA
| | - Ross P. Wohlgemuth
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Sarah E. Brashear
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | - Lucas R. Smith
- Department of Physiology, Neurobiology, and Behavior, University of California, Davis, Davis, CA, USA
| | | | - Theanne N. Griffith
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, USA
| |
Collapse
|
7
|
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
|
8
|
Ransdell JL, Carrasquillo Y, Bosch MK, Mellor RL, Ornitz DM, Nerbonne JM. Loss of Intracellular Fibroblast Growth Factor 14 (iFGF14) Increases the Excitability of Mature Hippocampal and Cortical Pyramidal Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592532. [PMID: 38746081 PMCID: PMC11092765 DOI: 10.1101/2024.05.04.592532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with progressive deficits in motor coordination and cognitive function. Mice ( Fgf14 -/- ) lacking iFGF14 display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to the loss of iFGF14-mediated regulation of the voltage-dependence of inactivation of the fast transient component of the voltage-gated Na + (Nav) current, I NaT . Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal and cortical pyramidal neurons. Current-clamp recordings from adult mouse hippocampal CA1 pyramidal neurons in acute in vitro slices, however, revealed that repetitive firing rates were higher in Fgf14 -/- , than in wild type (WT), cells. In addition, the waveforms of individual action potentials were altered in Fgf14 -/- hippocampal CA1 pyramidal neurons, and the loss of iFGF14 reduced the time delay between the initiation of axonal and somal action potentials. Voltage-clamp recordings revealed that the loss of iFGF14 altered the voltage-dependence of activation, but not inactivation, of I NaT in CA1 pyramidal neurons. Similar effects of the loss of iFGF14 on firing properties were evident in current-clamp recordings from layer 5 visual cortical pyramidal neurons. Additional experiments demonstrated that the loss of iFGF14 does not alter the distribution of anti-Nav1.6 or anti-ankyrin G immunofluorescence labeling intensity along the axon initial segments (AIS) of mature hippocampal CA1 or layer 5 visual cortical pyramidal neurons in situ . Taken together, the results demonstrate that, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, the loss of iFGF14 does not disrupt AIS architecture or Nav1.6 localization/distribution along the AIS of mature hippocampal (or cortical) pyramidal neurons in situ .
Collapse
|
9
|
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
|
10
|
Lyu H, Boßelmann CM, Johannesen KM, Koko M, Ortigoza-Escobar JD, Aguilera-Albesa S, Garcia-Navas Núñez D, Linnankivi T, Gaily E, van Ruiten HJA, Richardson R, Betzler C, Horvath G, Brilstra E, Geerdink N, Orsucci D, Tessa A, Gardella E, Fleszar Z, Schöls L, Lerche H, Møller RS, Liu Y. Clinical and electrophysiological features of SCN8A variants causing episodic or chronic ataxia. EBioMedicine 2023; 98:104855. [PMID: 38251463 PMCID: PMC10628346 DOI: 10.1016/j.ebiom.2023.104855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Variants in SCN8A are associated with a spectrum of epilepsies and neurodevelopmental disorders. Ataxia as a predominant symptom of SCN8A variation has not been well studied. We set out to investigate disease mechanisms and genotype-phenotype correlations of SCN8A-related ataxia. METHODS We collected genetic and electro-clinical data of ten individuals from nine unrelated families carrying novel SCN8A variants associated with chronic progressive or episodic ataxia. Electrophysiological characterizations of these variants were performed in ND7/23 cells and cultured neurons. FINDINGS Variants associated with chronic progressive ataxia either decreased Na+ current densities and shifted activation curves towards more depolarized potentials (p.Asn995Asp, p.Lys1498Glu and p.Trp1266Cys) or resulted in a premature stop codon (p.Trp937Ter). Three variants (p.Arg847Gln and biallelic p.Arg191Trp/p.Asp1525Tyr) were associated with episodic ataxia causing loss-of-function by decreasing Na+ current densities or a hyperpolarizing shift of the inactivation curve. Two additional episodic ataxia-associated variants caused mixed gain- and loss-of function effects in ND7/23 cells and were further examined in primary murine hippocampal neuronal cultures. Neuronal firing in excitatory neurons was increased by p.Arg1629His, but decreased by p.Glu1201Lys. Neuronal firing in inhibitory neurons was decreased for both variants. No functional effect was observed for p.Arg1913Trp. In four individuals, treatment with sodium channel blockers exacerbated symptoms. INTERPRETATION We identified episodic or chronic ataxia as predominant phenotypes caused by variants in SCN8A. Genotype-phenotype correlations revealed a more pronounced loss-of-function effect for variants causing chronic ataxia. Sodium channel blockers should be avoided under these conditions. FUNDING BMBF, DFG, the Italian Ministry of Health, University of Tuebingen.
Collapse
Affiliation(s)
- Hang Lyu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Christian M Boßelmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Katrine M Johannesen
- Department of Clinical Genetics, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark; Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark
| | - Mahmoud Koko
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Juan Dario Ortigoza-Escobar
- Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, CIBERER-ISCIII and European Reference Network for Rare Neurological Diseases (ERN-RND), Barcelona, Spain
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitario de Navarra, Pamplona, Spain; Navarrabiomed-Fundación Miguel Servet, Pamplona, Spain
| | | | - Tarja Linnankivi
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Eija Gaily
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Henriette J A van Ruiten
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Ruth Richardson
- Northern Genetics Service, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, UK
| | - Cornelia Betzler
- Institute for Rehabilitation, Transition and Palliation, Paracelsus Medical University, Salzburg, Austria; Specialist Center for Paediatric Neurology, Neuro-Rehabilitation and Epileptology, Schön Klinik Vogtareuth, Germany
| | - Gabriella Horvath
- Adult Metabolic Diseases Clinic, BC Children's Hospital, Vancouver, Canada
| | - Eva Brilstra
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niels Geerdink
- Department of Pediatrics, Rijnstate Hospital, Arnhem, the Netherlands
| | | | | | - Elena Gardella
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark; Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Zofia Fleszar
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ludger Schöls
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark; Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Yuanyuan Liu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
11
|
Rudolph S, Badura A, Lutzu S, Pathak SS, Thieme A, Verpeut JL, Wagner MJ, Yang YM, Fioravante D. Cognitive-Affective Functions of the Cerebellum. J Neurosci 2023; 43:7554-7564. [PMID: 37940582 PMCID: PMC10634583 DOI: 10.1523/jneurosci.1451-23.2023] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Stefano Lutzu
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, D-45147, Germany
| | - Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Mark J Wagner
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, Maryland 20814
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Diasynou Fioravante
- Center for Neuroscience, University of California-Davis, Davis, California 95618
- Department of Neurobiology, Physiology and Behavior, University of California-Davis, Davis, California 95618
| |
Collapse
|
12
|
Drouillas B, Brocard C, Zanella S, Bos R, Brocard F. Persistent Nav1.1 and Nav1.6 currents drive spinal locomotor functions through nonlinear dynamics. Cell Rep 2023; 42:113085. [PMID: 37665666 DOI: 10.1016/j.celrep.2023.113085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
Persistent sodium current (INaP) in the spinal locomotor network promotes two distinct nonlinear firing patterns: a self-sustained spiking triggered by a brief excitation in bistable motoneurons and bursting oscillations in interneurons of the central pattern generator (CPG). Here, we identify the NaV channels responsible for INaP and their role in motor behaviors. We report the axonal Nav1.6 as the main molecular player for INaP in lumbar motoneurons. The inhibition of Nav1.6, but not of Nav1.1, in motoneurons impairs INaP, bistability, postural tone, and locomotor performance. In interneurons of the rhythmogenic CPG region, both Nav1.6 and Nav1.1 equally mediate INaP. Inhibition of both channels is required to abolish oscillatory bursting activities and the locomotor rhythm. Overall, Nav1.6 plays a significant role both in posture and locomotion by governing INaP-dependent bistability in motoneurons and working in tandem with Nav1.1 to provide INaP-dependent rhythmogenic properties of the CPG.
Collapse
Affiliation(s)
- Benoît Drouillas
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Cécile Brocard
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Sébastien Zanella
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Rémi Bos
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France.
| |
Collapse
|
13
|
Rey Hipolito AG, van der Heijden ME, Sillitoe RV. Physiology of Dystonia: Animal Studies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 169:163-215. [PMID: 37482392 DOI: 10.1016/bs.irn.2023.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Dystonia is currently ranked as the third most prevalent motor disorder. It is typically characterized by involuntary muscle over- or co-contractions that can cause painful abnormal postures and jerky movements. Dystonia is a heterogenous disorder-across patients, dystonic symptoms vary in their severity, body distribution, temporal pattern, onset, and progression. There are also a growing number of genes that are associated with hereditary dystonia. In addition, multiple brain regions are associated with dystonic symptoms in both genetic and sporadic forms of the disease. The heterogeneity of dystonia has made it difficult to fully understand its underlying pathophysiology. However, the use of animal models has been used to uncover the complex circuit mechanisms that lead to dystonic behaviors. Here, we summarize findings from animal models harboring mutations in dystonia-associated genes and phenotypic animal models with overt dystonic motor signs resulting from spontaneous mutations, neural circuit perturbations, or pharmacological manipulations. Taken together, an emerging picture depicts dystonia as a result of brain-wide network dysfunction driven by basal ganglia and cerebellar dysfunction. In the basal ganglia, changes in dopaminergic, serotonergic, noradrenergic, and cholinergic signaling are found across different animal models. In the cerebellum, abnormal burst firing activity is observed in multiple dystonia models. We are now beginning to unveil the extent to which these structures mechanistically interact with each other. Such mechanisms inspire the use of pre-clinical animal models that will be used to design new therapies including drug treatments and brain stimulation.
Collapse
Affiliation(s)
- Alejandro G Rey Hipolito
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, United States
| | - Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, United States
| | - Roy V Sillitoe
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States; Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, United States; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, United States.
| |
Collapse
|
14
|
Dardiotis E, Skouras P, Varvarelis OP, Aloizou AM, Hernández AF, Liampas I, Rikos D, Dastamani M, Golokhvast KS, Bogdanos DP, Tsatsakis A, Siokas V, Mitsias PD, Hadjigeorgiou GM. Pesticides and tremor: An overview of association, mechanisms and confounders. ENVIRONMENTAL RESEARCH 2023; 229:115442. [PMID: 36758916 DOI: 10.1016/j.envres.2023.115442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 05/06/2023]
Abstract
Pesticides are a heterogeneous class of chemicals mainly used for the protection of crops from pests. Because of their very widespread use, acute or/and chronic exposure to these chemicals can lead to a plethora of sequelae inflicting diseases, many of which involve the nervous system. Tremor has been associated with pesticide exposure in human and animal studies. This review is aimed at assessing the studies currently available on the association between the various types of pesticides/insecticides and tremor, while also accounting for potential confounding factors. To our knowledge, this is the first coherent review on the subject. After appraising the available evidence, we call for more intensive research on this topic, as well as intonate the need of implementing future preventive measures to protect the exposed populations and to reduce potential disabilities and social drawbacks.
Collapse
Affiliation(s)
- Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece.
| | - Panagiotis Skouras
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Orfeas-Petros Varvarelis
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Antonio F Hernández
- Department of Legal Medicine and Toxicology, University of Granada School of Medicine, Granada, Spain; Health Research Institute of Granada (ibs.GRANADA), Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Ioannis Liampas
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Dimitrios Rikos
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Metaxia Dastamani
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Kirill S Golokhvast
- Siberian Federal Scientific Center of Agrobiotechnology RAS, Krasnoobsk, Russia, 630501
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, 71003, Heraklion, Crete, Greece
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Panayiotis D Mitsias
- Department of Neurology, School of Medicine, University of Crete, 71003, Heraklion, Greece; Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Georgios M Hadjigeorgiou
- Department of Neurology, University Hospital of Larissa Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece; Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
15
|
Johnson JP, Focken T, Khakh K, Tari PK, Dube C, Goodchild SJ, Andrez JC, Bankar G, Bogucki D, Burford K, Chang E, Chowdhury S, Dean R, de Boer G, Decker S, Dehnhardt C, Feng M, Gong W, Grimwood M, Hasan A, Hussainkhel A, Jia Q, Lee S, Li J, Lin S, Lindgren A, Lofstrand V, Mezeyova J, Namdari R, Nelkenbrecher K, Shuart NG, Sojo L, Sun S, Taron M, Waldbrook M, Weeratunge D, Wesolowski S, Williams A, Wilson M, Xie Z, Yoo R, Young C, Zenova A, Zhang W, Cutts AJ, Sherrington RP, Pimstone SN, Winquist R, Cohen CJ, Empfield JR. NBI-921352, a first-in-class, Na V1.6 selective, sodium channel inhibitor that prevents seizures in Scn8a gain-of-function mice, and wild-type mice and rats. eLife 2022; 11:72468. [PMID: 35234610 PMCID: PMC8903829 DOI: 10.7554/elife.72468] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
NBI-921352 (formerly XEN901) is a novel sodium channel inhibitor designed to specifically target NaV1.6 channels. Such a molecule provides a precision-medicine approach to target SCN8A-related epilepsy syndromes (SCN8A-RES), where gain-of-function (GoF) mutations lead to excess NaV1.6 sodium current, or other indications where NaV1.6 mediated hyper-excitability contributes to disease (Gardella and Møller, 2019; Johannesen et al., 2019; Veeramah et al., 2012). NBI-921352 is a potent inhibitor of NaV1.6 (IC500.051 µM), with exquisite selectivity over other sodium channel isoforms (selectivity ratios of 756 X for NaV1.1, 134 X for NaV1.2, 276 X for NaV1.7, and >583 Xfor NaV1.3, NaV1.4, and NaV1.5). NBI-921352is a state-dependent inhibitor, preferentially inhibiting inactivatedchannels. The state dependence leads to potent stabilization of inactivation, inhibiting NaV1.6 currents, including resurgent and persistent NaV1.6 currents, while sparing the closed/rested channels. The isoform-selective profile of NBI-921352 led to a robust inhibition of action-potential firing in glutamatergic excitatory pyramidal neurons, while sparing fast-spiking inhibitory interneurons, where NaV1.1 predominates. Oral administration of NBI-921352 prevented electrically induced seizures in a Scn8a GoF mouse,as well as in wild-type mouse and ratseizure models. NBI-921352 was effective in preventing seizures at lower brain and plasma concentrations than commonly prescribed sodium channel inhibitor anti-seizure medicines (ASMs) carbamazepine, phenytoin, and lacosamide. NBI-921352 waswell tolerated at higher multiples of the effective plasma and brain concentrations than those ASMs. NBI-921352 is entering phase II proof-of-concept trials for the treatment of SCN8A-developmental epileptic encephalopathy (SCN8A-DEE) and adult focal-onset seizures.
Collapse
Affiliation(s)
- J P Johnson
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Thilo Focken
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Kuldip Khakh
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Celine Dube
- In Vivo Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | | | - Girish Bankar
- In Vivo Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - David Bogucki
- Chemistry, Medipure Pharmaceuticals, Burnaby BC, Canada
| | | | - Elaine Chang
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Richard Dean
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Gina de Boer
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Shannon Decker
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Mandy Feng
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Wei Gong
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Abid Hasan
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Qi Jia
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Stephanie Lee
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Jenny Li
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Sophia Lin
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Andrea Lindgren
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Janette Mezeyova
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Rostam Namdari
- Translational Drug Development, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | | | - Luis Sojo
- Compound Properties, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Shaoyi Sun
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Matthew Taron
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Diana Weeratunge
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | - Aaron Williams
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Michael Wilson
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Zhiwei Xie
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Rhena Yoo
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Clint Young
- In Vitro Biology, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Alla Zenova
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Wei Zhang
- Chemistry, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | - Alison J Cutts
- Scientific Affairs, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | | | | | | - Charles J Cohen
- Executive Team, Xenon Pharmaceuticals, Inc., Burnaby BC, Canada
| | | |
Collapse
|
16
|
Yang X, Yin H, Wang X, Sun Y, Bian X, Zhang G, Li A, Cao A, Li B, Ebrahimi-Fakhari D, Yang Z, Meisler MH, Liu Q. Social Deficits and Cerebellar Degeneration in Purkinje Cell Scn8a Knockout Mice. Front Mol Neurosci 2022; 15:822129. [PMID: 35557557 PMCID: PMC9087741 DOI: 10.3389/fnmol.2022.822129] [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: 11/25/2021] [Accepted: 02/18/2022] [Indexed: 11/23/2022] Open
Abstract
Mutations in the SCN8A gene encoding the voltage-gated sodium channel α-subunit Nav1. 6 have been reported in individuals with epilepsy, intellectual disability and features of autism spectrum disorder. SCN8A is widely expressed in the central nervous system, including the cerebellum. Cerebellar dysfunction has been implicated in autism spectrum disorder. We investigated conditional Scn8a knockout mice under C57BL/6J strain background that specifically lack Scn8a expression in cerebellar Purkinje cells (Scn8a flox/flox , L7Cre + mice). Cerebellar morphology was analyzed by immunohistochemistry and MR imaging. Mice were subjected to a battery of behavioral tests including the accelerating rotarod, open field, elevated plus maze, light-dark transition box, three chambers, male-female interaction, social olfaction, and water T-maze tests. Patch clamp recordings were used to evaluate evoked action potentials in Purkinje cells. Behavioral phenotyping demonstrated that Scn8a flox/flox , L7Cre + mice have impaired social interaction, motor learning and reversal learning as well as increased repetitive behavior and anxiety-like behaviors. By 5 months of age, Scn8a flox/flox , L7Cre + mice began to exhibit cerebellar Purkinje cell loss and reduced molecular thickness. At 9 months of age, Scn8a flox/flox , L7Cre + mice exhibited decreased cerebellar size and a reduced number of cerebellar Purkinje cells more profoundly, with evidence of additional neurodegeneration in the molecular layer and deep cerebellar nuclei. Purkinje cells in Scn8a flox/flox , L7Cre + mice exhibited reduced repetitive firing. Taken together, our experiments indicated that loss of Scn8a expression in cerebellar Purkinje cells leads to cerebellar degeneration and several ASD-related behaviors. Our study demonstrated the specific contribution of loss of Scn8a in cerebellar Purkinje cells to behavioral deficits characteristic of ASD. However, it should be noted that our observed effects reported here are specific to the C57BL/6 genome type.
Collapse
Affiliation(s)
- Xiaofan Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hongqiang Yin
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, China.,Department of Operational Medicine, Tianjin Institute of Environmental & Operational Medicine, Tianjin, China
| | - Xiaojing Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yueqing Sun
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xianli Bian
- Department of Neurology, Second Hospital of Shandong University, Jinan, China
| | - Gaorui Zhang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Anning Li
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Aihua Cao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Baomin Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Zhuo Yang
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, China
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States.,Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Qiji Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China.,Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, China
| |
Collapse
|
17
|
Tyszkiewicz C, Pardo ID, Ritenour HN, Liu CN, Somps C. Increases in GFAP immunoreactive astrocytes in the cerebellar molecular layer of young adult CBA/J mice. Lab Anim Res 2021; 37:24. [PMID: 34454633 PMCID: PMC8400896 DOI: 10.1186/s42826-021-00100-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
Background CBA/J mice are standard experimental animals in auditory studies, and age-related changes in auditory pathways are well documented. However, changes in locomotion-related brain regions have not been systematically explored. Results We showed an increase in immunoreactivity for glial fibrillary acidic protein (GFAP) in the cerebellar molecular layer associated with Purkinje cells in mice at 24 weeks of age but not in the younger mice. Increased GFAP immunoreactivity appeared in the form of clusters and distributed multifocally consistent with hyperplasia of astrocytes that were occasionally associated with Purkinje cell degeneration. Three out of 12 animals at 16 and 24 weeks of age exhibited pre-convulsive clinical signs. Two of these 3 animals also showed increased GFAP immunoreactivity in the cerebellum. Rotarod behavioral assessments indicated decreased performance at 24 weeks of age. Conclusions These results suggest minimal to mild reactive astrocytosis likely associated with Purkinje cell degeneration in the cerebellum at 24 weeks of age in CBA/J mice. These findings should be taken into consideration prior to using this mouse strain for studying neuroinflammation or aging. Supplementary Information The online version contains supplementary material available at 10.1186/s42826-021-00100-5.
Collapse
Affiliation(s)
- Cheryl Tyszkiewicz
- Comparative Medicine, Worldwide Research, Development and Medical, Pfizer Inc, MS 8274-1359, PGRD, Eastern Point Road, Groton, CT, 06340, USA
| | - Ingrid D Pardo
- Global Pathology and Investigative Toxicology, Pfizer Inc, Groton, CT, 06340, USA
| | - Hayley N Ritenour
- Global Pathology and Investigative Toxicology, Pfizer Inc, Groton, CT, 06340, USA
| | - Chang-Ning Liu
- Comparative Medicine, Worldwide Research, Development and Medical, Pfizer Inc, MS 8274-1359, PGRD, Eastern Point Road, Groton, CT, 06340, USA.
| | - Chris Somps
- Global Pathology and Investigative Toxicology, Pfizer Inc, Groton, CT, 06340, USA
| |
Collapse
|
18
|
Zybura A, Hudmon A, Cummins TR. Distinctive Properties and Powerful Neuromodulation of Na v1.6 Sodium Channels Regulates Neuronal Excitability. Cells 2021; 10:1595. [PMID: 34202119 PMCID: PMC8307729 DOI: 10.3390/cells10071595] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) are critical determinants of cellular excitability. These ion channels exist as large heteromultimeric structures and their activity is tightly controlled. In neurons, the isoform Nav1.6 is highly enriched at the axon initial segment and nodes, making it critical for the initiation and propagation of neuronal impulses. Changes in Nav1.6 expression and function profoundly impact the input-output properties of neurons in normal and pathological conditions. While mutations in Nav1.6 may cause channel dysfunction, aberrant changes may also be the result of complex modes of regulation, including various protein-protein interactions and post-translational modifications, which can alter membrane excitability and neuronal firing properties. Despite decades of research, the complexities of Nav1.6 modulation in health and disease are still being determined. While some modulatory mechanisms have similar effects on other Nav isoforms, others are isoform-specific. Additionally, considerable progress has been made toward understanding how individual protein interactions and/or modifications affect Nav1.6 function. However, there is still more to be learned about how these different modes of modulation interact. Here, we examine the role of Nav1.6 in neuronal function and provide a thorough review of this channel's complex regulatory mechanisms and how they may contribute to neuromodulation.
Collapse
Affiliation(s)
- Agnes Zybura
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Andy Hudmon
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA;
| | - Theodore R. Cummins
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
19
|
Praticò A, Gulizia C, Gangi G, Oliva C, Romano C, Marino S, Polizzi A, Ruggieri M, Falsaperla R. SCN8A and Its Related Epileptic Phenotypes. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1729142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractSodium channelopathies are among the most common single-gene causes of epilepsy and have been considered model disorders for the study of genetic epilepsies. Epilepsies due to SCN8A pathogenic variants can present with a broad range of phenotypes varying from a severe epileptic encephalopathy with multiple types of drug-resistant seizure to neurodevelopmental delay, mental retardation, and electroencephalogram (EEG) findings of multifocal spike and waves (mostly in the temporal/parietal/occipital areas). In rare cases, benign familial infantile seizures and developmental delay with/without ataxia have been reported. A first-level, specific SCN8A Sanger's sequencing, although available, is rarely performed because the clinical phenotype is not strictly characteristic and several overlaps with other genetic epilepsies may occur. Given its indistinctive phenotype, diagnosis is usually performed through a specific gene panel for epileptic encephalopathies, early epilepsies, or genetic epilepsy in general, or through whole exome sequencing (WES) and more rarely through whole genome sequencing (WGS). Mutations in SCN8A occur as an autosomal dominant trait. The great majority of individuals diagnosed with SCN8A epilepsy do not have an affected parent, because usually SCN8A patients do not reproduce, and mutations are inherited as a “de novo” trait. In rare cases, SCN8A mutations may be inherited in the setting of parental germline mosaicism. SCN8A-related epilepsies have not shown a clear genotype–phenotype correlation, the same variants have been described with different clinical expressivity and this could be due to other genetic factors or to interacting environmental factors. There is no standardized treatment for SCN8A-related epilepsy because of the rarity of the disease and the unavailability of specific, targeted drugs. Treatment is based mainly on antiepileptic drugs which include classic wide-spectrum drugs such as valproic acid, levetiracetam, and lamotrigine. Sodium-channel blockers (phenytoin, carbamazepine, oxcarbazepine, and lamotrigine) have shown appreciable results in terms of seizure reduction, in particular, in patients presenting gain-of-function mutations. Nowadays, new potentially transformative gene therapy treatment approaches are currently being explored, allowing in the next future, a precision-based treatment directed against the gene defect and protein alterations.
Collapse
Affiliation(s)
- Andrea Praticò
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | - Carmela Gulizia
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Gloria Gangi
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Claudia Oliva
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Simona Marino
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| | - Agata Polizzi
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
| | - Martino Ruggieri
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| |
Collapse
|
20
|
SCN8A Encephalopathy: Case Report and Literature Review. Neurol Int 2021; 13:143-150. [PMID: 33915942 PMCID: PMC8167728 DOI: 10.3390/neurolint13020014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Epileptic encephalopathy is a condition resulting from extreme forms of intractable childhood epilepsy. The disease can cause severe delays in cognitive, sensory, and motor function development, in addition to being fatal in some cases. Missense mutations of SCN8A, which encodes Nav1.6, one of the main voltage-gated sodium channel subunits in neurons and muscles, have been linked to early infantile SCN8A encephalopathy. Herein, we report the case of a 5-month-old girl with SCN8A encephalopathy with a novel missense mutation. Apart from intractable seizures and autistic phenotypes, the results of blood and biochemical tests, electroencephalogram (EEG) results, and brain magnetic resonance imaging (MRI) results were all normal. As the phenotypes caused by these mutations cannot be identified by any clinical, neuroimaging, or electrophysiological features, genetic sequencing should be considered to identify the underlying genetic causes. Although phenytoin is recommended as a last-resort treatment for SCN8A encephalopathy, the administration of the oxcarbazepine, instead of phenytoin, mitigated this patient's intractable seizures.
Collapse
|
21
|
Abstract
The voltage-gated sodium channel α-subunit genes comprise a highly conserved gene family. Mutations of three of these genes, SCN1A, SCN2A and SCN8A, are responsible for a significant burden of neurological disease. Recent progress in identification and functional characterization of patient variants is generating new insights and novel approaches to therapy for these devastating disorders. Here we review the basic elements of sodium channel function that are used to characterize patient variants. We summarize a large body of work using global and conditional mouse mutants to characterize the in vivo roles of these channels. We provide an overview of the neurological disorders associated with mutations of the human genes and examples of the effects of patient mutations on channel function. Finally, we highlight therapeutic interventions that are emerging from new insights into mechanisms of sodium channelopathies.
Collapse
|
22
|
Cook AA, Fields E, Watt AJ. Losing the Beat: Contribution of Purkinje Cell Firing Dysfunction to Disease, and Its Reversal. Neuroscience 2020; 462:247-261. [PMID: 32554108 DOI: 10.1016/j.neuroscience.2020.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023]
Abstract
The cerebellum is a brain structure that is highly interconnected with other brain regions. There are many contributing factors to cerebellar-related brain disease, such as altered afferent input, local connectivity, and/or cerebellar output. Purkinje cells (PC) are the principle cells of the cerebellar cortex, and fire intrinsically; that is, they fire spontaneous action potentials at high frequencies. This review paper focuses on PC intrinsic firing activity, which is altered in multiple neurological diseases, including ataxia, Huntington Disease (HD) and autism spectrum disorder (ASD). Notably, there are several cases where interventions that restore or rescue PC intrinsic activity also improve impaired behavior in these mouse models of disease. These findings suggest that rescuing PC firing deficits themselves may be sufficient to improve impairment in cerebellar-related behavior in disease. We propose that restoring PC intrinsic firing represents a good target for drug development that might be of therapeutic use for several disorders.
Collapse
Affiliation(s)
- Anna A Cook
- Department of Biology, McGill University, Montreal, Canada
| | - Eviatar Fields
- Department of Biology, McGill University, Montreal, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Alanna J Watt
- Department of Biology, McGill University, Montreal, Canada.
| |
Collapse
|
23
|
Essential tremor pathology: neurodegeneration and reorganization of neuronal connections. Nat Rev Neurol 2020; 16:69-83. [PMID: 31959938 DOI: 10.1038/s41582-019-0302-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2019] [Indexed: 01/26/2023]
Abstract
Essential tremor (ET) is the most common tremor disorder globally and is characterized by kinetic tremor of the upper limbs, although other clinical features can also occur. Postmortem studies are a particularly important avenue for advancing our understanding of the pathogenesis of ET; however, until recently, the number of such studies has been limited. Several recent postmortem studies have made important contributions to our understanding of the pathological changes that take place in ET. These studies identified abnormalities in the cerebellum, which primarily affected Purkinje cells (PCs), basket cells and climbing fibres, in individuals with ET. We suggest that some of these pathological changes (for example, focal PC axonal swellings, swellings in and regression of the PC dendritic arbor and PC death) are likely to be primary and degenerative. By contrast, other changes, such as an increase in PC recurrent axonal collateral formation and hypertrophy of GABAergic basket cell axonal processes, could be compensatory responses to restore cerebellar GABAergic tone and cerebellar cortical inhibitory efficacy. Such compensatory responses are likely to be insufficient, enabling the disease to progress. Here, we review the results of recent postmortem studies of ET and attempt to place these findings into an anatomical-physiological disease model.
Collapse
|
24
|
Alexander CJ, Hammer JA. An Improved Method for Differentiating Mouse Embryonic Stem Cells into Cerebellar Purkinje Neurons. THE CEREBELLUM 2019; 18:406-421. [PMID: 30729383 DOI: 10.1007/s12311-019-1007-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
While mixed primary cerebellar cultures prepared from embryonic tissue have proven valuable for dissecting structure-function relationships in cerebellar Purkinje neurons (PNs), this technique is technically challenging and often yields few cells. Recently, mouse embryonic stem cells (mESCs) have been successfully differentiated into PNs, although the published methods are very challenging as well. The focus of this study was to simplify the differentiation of mESCs into PNs. Using a recently described neural differentiation media, we generate monolayers of neural progenitor cells from mESCs and differentiate them into PN precursors using specific extrinsic factors. These PN precursors are then differentiated into mature PNs by co-culturing them with granule neuron (GN) precursors also derived from neural progenitors using different extrinsic factors. The morphology of mESC-derived PNs is indistinguishable from PNs grown in primary culture in terms of gross morphology, spine length, and spine density. Furthermore, mESC-derived PNs express Calbindin D28K, IP3R1, IRBIT, PLCβ4, PSD93, and myosin IIB-B2, all of which are either PN-specific or highly expressed in PNs. Moreover, we show that mESC-derived PNs form synapses with GN-like cells as in primary culture, express proteins driven by the PN-specific promoter Pcp2/L7, and exhibit the defect in spine ER inheritance seen in PNs isolated from dilute-lethal (myosin Va-null) mice when expressing a Pcp2/L7-driven miRNA directed against myosin Va. Finally, we define a novel extracellular matrix formulation that reproducibly yields monolayer cultures conducive for high-resolution imaging. Our improved method for differentiating mESCs into PNs should facilitate the dissection of molecular mechanisms and disease phenotypes in PNs.
Collapse
Affiliation(s)
- Christopher J Alexander
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - John A Hammer
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
25
|
White HV, Brown ST, Bozza TC, Raman IM. Effects of FGF14 and Na Vβ4 deletion on transient and resurgent Na current in cerebellar Purkinje neurons. J Gen Physiol 2019; 151:1300-1318. [PMID: 31558566 PMCID: PMC6829560 DOI: 10.1085/jgp.201912390] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/03/2019] [Indexed: 11/20/2022] Open
Abstract
Voltage-gated Na channels of Purkinje cells are specialized to maintain high availability during high-frequency repetitive firing. They enter fast-inactivated states relatively slowly and undergo a voltage-dependent open-channel block by an intracellular protein (or proteins) that prevents stable fast inactivation and generates resurgent Na current. These properties depend on the pore-forming α subunits, as well as modulatory subunits within the Na channel complex. The identity of the factors responsible for open-channel block remains a question. Here we investigate the effects of genetic mutation of two Na channel auxiliary subunits highly expressed in Purkinje cells, NaVβ4 and FGF14, on modulating Na channel blocked as well as inactivated states. We find that although both NaVβ4 and the FGF14 splice variant FGF14-1a contain sequences that can generate resurgent-like currents when applied to Na channels in peptide form, deletion of either protein, or both proteins simultaneously, does not eliminate resurgent current in acutely dissociated Purkinje cell bodies. Loss of FGF14 expression does, however, reduce resurgent current amplitude and leads to an acceleration and stabilization of inactivation that is not reversed by application of the site-3 toxin, anemone toxin II (ATX). Tetrodotoxin (TTX) sensitivity is higher for resurgent than transient components of Na current, and loss of FGF14 preferentially affects a highly TTX-sensitive subset of Purkinje α subunits. The data suggest that NaV1.6 channels, which are known to generate the majority of Purkinje cell resurgent current, bind TTX with high affinity and are modulated by FGF14 to facilitate open-channel block.
Collapse
Affiliation(s)
- Hayley V White
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Spencer T Brown
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Thomas C Bozza
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, IL .,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| |
Collapse
|
26
|
Venugopal S, Seki S, Terman DH, Pantazis A, Olcese R, Wiedau-Pazos M, Chandler SH. Resurgent Na+ Current Offers Noise Modulation in Bursting Neurons. PLoS Comput Biol 2019; 15:e1007154. [PMID: 31226124 PMCID: PMC6608983 DOI: 10.1371/journal.pcbi.1007154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 07/03/2019] [Accepted: 06/04/2019] [Indexed: 01/20/2023] Open
Abstract
Neurons utilize bursts of action potentials as an efficient and reliable way to encode information. It is likely that the intrinsic membrane properties of neurons involved in burst generation may also participate in preserving its temporal features. Here we examined the contribution of the persistent and resurgent components of voltage-gated Na+ currents in modulating the burst discharge in sensory neurons. Using mathematical modeling, theory and dynamic-clamp electrophysiology, we show that, distinct from the persistent Na+ component which is important for membrane resonance and burst generation, the resurgent Na+ can help stabilize burst timing features including the duration and intervals. Moreover, such a physiological role for the resurgent Na+ offered noise tolerance and preserved the regularity of burst patterns. Model analysis further predicted a negative feedback loop between the persistent and resurgent gating variables which mediate such gain in burst stability. These results highlight a novel role for the voltage-gated resurgent Na+ component in moderating the entropy of burst-encoded neural information.
Collapse
Affiliation(s)
- Sharmila Venugopal
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Soju Seki
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - David H Terman
- Department of Mathematics, The Ohio State University, Columbus, OH, United States of America
| | - Antonios Pantazis
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America.,Division of Neurobiology Department of Clinical and Experimental Medicine (IKE) and Wallenberg Center for Molecular Medicine Linköping University 581 83 Linköping Sweden
| | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Martina Wiedau-Pazos
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Scott H Chandler
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States of America
| |
Collapse
|
27
|
Deletion of Class II ADP-Ribosylation Factors in Mice Causes Tremor by the Nav1.6 Loss in Cerebellar Purkinje Cell Axon Initial Segments. J Neurosci 2019; 39:6339-6353. [PMID: 31201232 DOI: 10.1523/jneurosci.2002-18.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 05/14/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022] Open
Abstract
ADP-ribosylation factors (ARFs) are a family of small monomeric GTPases comprising six members categorized into three classes: class I (ARF1, 2, and 3), class II (ARF4 and 5), and class III (ARF6). In contrast to class I and III ARFs, which are the key regulators in vesicular membrane trafficking, the cellular function of class II ARFs remains unclear. In the present study, we generated class II ARF-deficient mice and found that ARF4+/-/ARF5-/- mice exhibited essential tremor (ET)-like behaviors. In vivo electrophysiological recordings revealed that ARF4+/-/ARF5-/- mice of both sexes exhibited abnormal brain activity when moving, raising the possibility of abnormal cerebellar excitability. Slice patch-clamp experiments demonstrated the reduced excitability of the cerebellar Purkinje cells (PCs) in ARF4+/-/ARF5-/- mice. Immunohistochemical and electrophysiological analyses revealed a severe and selective decrease of pore-forming voltage-dependent Na+ channel subunit Nav1.6, important for maintaining repetitive action potential firing, in the axon initial segment (AIS) of PCs. Importantly, this decrease in Nav1.6 protein localized in the AIS and the consequent tremors in ARF4+/-/ARF5-/- mice could be alleviated by the PC-specific expression of ARF5 using adeno-associated virus vectors. Together, our data demonstrate that the decreased expression of the class II ARF proteins in ARF4+/-/ARF5-/- mice, leading to a haploinsufficiency of ARF4 in the absence of ARF5, impairs the localization of Nav1.6 to the AIS and hence reduces the membrane excitability in PCs, resulting in the ET-like movement disorder. We suggest that class II ARFs function in localizing specific proteins, such as Nav1.6, to the AIS.SIGNIFICANCE STATEMENT We found that decreasing the expression of class II ARF proteins, through the generation of ARF4+/-/ARF5-/- mice, impairs Nav1.6 distribution to the axon initial segment (AIS) of cerebellar Purkinje cells (PCs), thereby resulting in the impairment of action potential firing of PCs. The ARF4+/-/ARF5-/- mutant mice exhibited movement-associated essential tremor (ET)-like behavior with pharmacological profiles similar to those in ET patients. The exogenous expression of ARF5 reduced the tremor phenotype and restored the localization of Nav1.6 immunoreactivity to the AIS in ARF4+/-/ARF5-/- mice. Thus, our results suggest that class II ARFs are involved in the localization of Nav1.6 to the AISs in cerebellar PCs and that the reduction of class II ARF activity leads to ET-like movement disorder.
Collapse
|
28
|
Johannesen KM, Gardella E, Encinas AC, Lehesjoki A, Linnankivi T, Petersen MB, Lund ICB, Blichfeldt S, Miranda MJ, Pal DK, Lascelles K, Procopis P, Orsini A, Bonuccelli A, Giacomini T, Helbig I, Fenger CD, Sisodiya SM, Hernandez‐Hernandez L, Krithika S, Rumple M, Masnada S, Valente M, Cereda C, Giordano L, Accorsi P, Bürki SE, Mancardi M, Korff C, Guerrini R, Spiczak S, Hoffman‐Zacharska D, Mazurczak T, Coppola A, Buono S, Vecchi M, Hammer MF, Varesio C, Veggiotti P, Lal D, Brünger T, Zara F, Striano P, Rubboli G, Møller RS. The spectrum of intermediate
SCN
8A
‐related epilepsy. Epilepsia 2019; 60:830-844. [DOI: 10.1111/epi.14705] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/07/2019] [Accepted: 03/07/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Katrine M. Johannesen
- Department of Epilepsy Genetics and Personalized TreatmentDanish Epilepsy Center Filadelfia Dianalund Denmark
- Institute for Regional Health ServicesUniversity of Southern Denmark Odense Denmark
| | - Elena Gardella
- Department of Epilepsy Genetics and Personalized TreatmentDanish Epilepsy Center Filadelfia Dianalund Denmark
- Institute for Regional Health ServicesUniversity of Southern Denmark Odense Denmark
| | - Alejandra C. Encinas
- Graduate Interdisciplinary Program of GeneticsUniversity of Arizona Tucson Arizona
| | - Anna‐Elina Lehesjoki
- Folkhälsan Research Center Helsinki Finland
- Research Programs Unit, Molecular Neurology and MedicumUniversity of Helsinki Helsinki Finland
| | - Tarja Linnankivi
- Department of Child NeurologyChildren's HospitalUniversity of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Michael B. Petersen
- Department of Clinical GeneticsAalborg University Hospital Aalborg Denmark
- Department of Clinical MedicineAalborg University Aalborg Denmark
| | | | | | | | - Deb K. Pal
- Department of Basic and Clinical NeuroscienceInstitute of Psychiatry, Psychology, and NeuroscienceKing's College London London UK
- King's College Hospital London UK
- Evelina London Children's Hospital London UK
- Medical Research Council Centre for Neurodevelopmental DisordersKing's College London UK
| | - Karine Lascelles
- Department of Basic and Clinical NeuroscienceInstitute of Psychiatry, Psychology, and NeuroscienceKing's College London London UK
| | - Peter Procopis
- Children's Hospital Westmead, Sydney New South Wales Australia
- Discipline of Child and Adolescent HealthSydney Medical SchoolUniversity of Sydney Sydney New South Wales Australia
| | | | - Alice Bonuccelli
- Pediatric NeurologyPediatric ClinicUniversity of Pisa Pisa Italy
| | - Thea Giacomini
- Child Neuropsychiatry UnitDepartment of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Children's SciencesGiannina Gaslini InstituteUniversity of Genoa Genoa Italy
| | - Ingo Helbig
- Department of NeuropediatricsUniversity Medical Center Schleswig Holstein Kiel Germany
- Division of NeurologyChildren's Hospital of Philadelphia Philadelphia Pennsylvania
| | - Christina D. Fenger
- Department of Epilepsy Genetics and Personalized TreatmentDanish Epilepsy Center Filadelfia Dianalund Denmark
| | - Sanjay M. Sisodiya
- Department of Clinical and Experimental EpilepsyUniversity College London Institute of Neurology London UK
- Chalfont Centre for Epilepsy Bucks UK
| | - Laura Hernandez‐Hernandez
- Department of Clinical and Experimental EpilepsyUniversity College London Institute of Neurology London UK
- Chalfont Centre for Epilepsy Bucks UK
| | - Sundararaman Krithika
- Department of Clinical and Experimental EpilepsyUniversity College London Institute of Neurology London UK
- Chalfont Centre for Epilepsy Bucks UK
| | - Melissa Rumple
- Pediatric NeurologyBanner Children's Specialists Glendale Arizona
| | - Silvia Masnada
- Department of Brain and Behavioral SciencesUniversity of Pavia Pavia Italy
| | - Marialuisa Valente
- Genomic and Postgenomic CenterScientific Institute for Research and Healthcare (IRCCS) Mondino Foundation Pavia Italy
| | - Cristina Cereda
- Genomic and Postgenomic CenterScientific Institute for Research and Healthcare (IRCCS) Mondino Foundation Pavia Italy
| | - Lucio Giordano
- Child Neurology and Psychiatry UnitCivilian Hospital Brescia Italy
| | - Patrizia Accorsi
- Child Neurology and Psychiatry UnitCivilian Hospital Brescia Italy
| | - Sarah E. Bürki
- Department of PediatricsDivision of Child NeurologyUniversity Children's Hospital BernUniversity of Bern Bern Switzerland
| | - Margherita Mancardi
- Unit of Child NeuropsychiatryEpilepsy CenterDepartment of Clinical and Surgical Neuroscience and RehabilitationGiannina Gaslini Institute Genoa Italy
| | - Christian Korff
- Child Neurology UnitUniversity Children's Hospital Geneva Switzerland
| | - Renzo Guerrini
- Neuroscience DepartmentChildren's Hospital Anna Meyer, University of Florence Florence Italy
| | - Sarah Spiczak
- Department of NeuropediatricsChristian Albrecht University Kiel Germany
- Northern German Epilepsy Center for Children and Adolescents Schwentinental Germany
| | | | - Tomasz Mazurczak
- Department of Neurology of Children and AdolescentsInstitute of Mother and Child Warsaw Poland
| | - Antonietta Coppola
- Department of Neuroscience and Reproductive and Odontostomatological SciencesFederico II University Naples Italy
| | - Salvatore Buono
- Neurology DivisionHospital of National Relevance (AORN), Santobono Pausilipon Naples Italy
| | - Marilena Vecchi
- Pediatric Clinic, Hospital CompanyUniversity of Padua Padua Italy
| | - Michael F. Hammer
- University of Arizona Genetic CoreUniversity of Arizona Tucson Arizona
| | - Costanza Varesio
- Brain and Behavior DepartmentUniversity of Pavia Pavia Italy
- Child and Adolescence Neurology DepartmentIRCCS C. Mondino National Neurological Institute Pavia Italy
| | - Pierangelo Veggiotti
- Department of Child NeurologyV. Buzzi Children's Hospital Milan Italy
- L. Sacco Department of Biomedical and Clinical SciencesUniversity of Milan Milan Italy
| | - Dennis Lal
- Epilepsy CenterNeurological InstituteCleveland Clinic Cleveland Ohio
- Genomic Medicine InstituteLerner Research Institute Cleveland Clinic Cleveland Ohio
- Stanley Center for Psychiatric ResearchBroad Institute of Massachusetts Institute of Technology and Harvard Cambridge Massachusetts
- Analytic and Translational Genetics UnitMassachusetts General Hospital Boston Massachusetts
- Cologne Center for GenomicsUniversity of Cologne Cologne Germany
| | - Tobias Brünger
- Cologne Center for GenomicsUniversity of Cologne Cologne Germany
| | - Federico Zara
- Laboratory of Neurogenetics and NeuroscienceDepartment of Head‐Neck and NeuroscienceGiannina Gaslini Institute Genoa Italy
| | - Pasquale Striano
- Pediatric NeurologyPediatric ClinicUniversity of Studies of Pisa Pisa Italy
| | - Guido Rubboli
- Department of Epilepsy Genetics and Personalized TreatmentDanish Epilepsy Center Filadelfia Dianalund Denmark
- University of Copenhagen Copenhagen Denmark
| | - Rikke S. Møller
- Department of Epilepsy Genetics and Personalized TreatmentDanish Epilepsy Center Filadelfia Dianalund Denmark
- Institute for Regional Health ServicesUniversity of Southern Denmark Odense Denmark
| |
Collapse
|
29
|
Mazzonetto PC, Ariza CB, Ocanha SG, de Souza TA, Ko GM, Menck CFM, Massironi SMG, Porcionatto MA. Mutation in NADPH oxidase 3 (NOX3) impairs SHH signaling and increases cerebellar neural stem/progenitor cell proliferation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1502-1515. [PMID: 30853403 DOI: 10.1016/j.bbadis.2019.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 01/03/2023]
Abstract
Abnormalities in cerebellar structure and function may cause ataxia, a neurological dysfunction of motor coordination. In the course of the present study, we characterized a mutant mouse lineage with an ataxia-like phenotype. We localized the mutation on chromosome 17 and mapped it to position 1534 of the Nox3 gene, resulting in p.Asn64Tyr change. The primary defect observed in Nox3eqlb mice was increased proliferation of cerebellar granule cell precursors (GCPs). cDNA microarray comparing Nox3eqlb and BALB/c neonatal cerebellum revealed changes in the expression of genes involved in the control of cell proliferation. Nox3eqlb GCPs and NSC produce higher amounts of reactive oxygen species (ROS) and upregulate the expression of SHH target genes, such as Gli1-3 and Ccnd1 (CyclinD1). We hypothesize that this new mutation is responsible for an increase in proliferation via stimulation of the SHH pathway. We suggest this mutant mouse lineage as a new model to investigate the role of ROS in neuronal precursor cell proliferation.
Collapse
Affiliation(s)
- P C Mazzonetto
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - C B Ariza
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil; Department of General Pathology, Center of Biological Sciences, Universidade Estadual de Londrina (UEL), Brazil
| | - S G Ocanha
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - T A de Souza
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - G M Ko
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - C F M Menck
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - S M G Massironi
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - M A Porcionatto
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil.
| |
Collapse
|
30
|
Meredith FL, Rennie KJ. Regional and Developmental Differences in Na + Currents in Vestibular Primary Afferent Neurons. Front Cell Neurosci 2018; 12:423. [PMID: 30487736 PMCID: PMC6246661 DOI: 10.3389/fncel.2018.00423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/04/2023] Open
Abstract
The vestibular system relays information about head position via afferent nerve fibers to the brain in the form of action potentials. Voltage-gated Na+ channels in vestibular afferents drive the initiation and propagation of action potentials, but their expression during postnatal development and their contributions to firing in diverse mature afferent populations are unknown. Electrophysiological techniques were used to determine Na+ channel subunit types in vestibular calyx-bearing afferents at different stages of postnatal development. We used whole cell patch clamp recordings in thin slices of gerbil crista neuroepithelium to investigate Na+ channels and firing patterns in central zone (CZ) and peripheral zone (PZ) afferents. PZ afferents are exclusively dimorphic, innervating type I and type II hair cells, whereas CZ afferents can form dimorphs or calyx-only terminals which innervate type I hair cells alone. All afferents expressed tetrodotoxin (TTX)-sensitive Na+ currents, but TTX-sensitivity varied with age. During the fourth postnatal week, 200–300 nM TTX completely blocked sodium currents in PZ and CZ calyces. By contrast, in immature calyces [postnatal day (P) 5–11], a small component of peak sodium current remained in 200 nM TTX. Application of 1 μM TTX, or Jingzhaotoxin-III plus 200 nM TTX, abolished sodium current in immature calyces, suggesting the transient expression of voltage-gated sodium channel 1.5 (Nav1.5) during development. A similar TTX-insensitive current was found in early postnatal crista hair cells (P5–9) and constituted approximately one third of the total sodium current. The Nav1.6 channel blocker, 4,9-anhydrotetrodotoxin, reduced a component of sodium current in immature and mature calyces. At 100 nM 4,9-anhydrotetrodotoxin, peak sodium current was reduced on average by 20% in P5–14 calyces, by 37% in mature dimorphic PZ calyces, but by less than 15% in mature CZ calyx-only terminals. In mature PZ calyces, action potentials became shorter and broader in the presence of 4,9-anhydrotetrodotoxin implicating a role for Nav1.6 channels in firing in dimorphic afferents.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
31
|
Ransdell JL, Nerbonne JM. Voltage-gated sodium currents in cerebellar Purkinje neurons: functional and molecular diversity. Cell Mol Life Sci 2018; 75:3495-3505. [PMID: 29982847 PMCID: PMC6123253 DOI: 10.1007/s00018-018-2868-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
Purkinje neurons, the sole output of the cerebellar cortex, deliver GABA-mediated inhibition to the deep cerebellar nuclei. To subserve this critical function, Purkinje neurons fire repetitively, and at high frequencies, features that have been linked to the unique properties of the voltage-gated sodium (Nav) channels expressed. In addition to the rapidly activating and inactivating, or transient, component of the Nav current (INaT) present in many types of central and peripheral neurons, Purkinje neurons, also expresses persistent (INaP) and resurgent (INaR) Nav currents. Considerable progress has been made in detailing the biophysical properties and identifying the molecular determinants of these discrete Nav current components, as well as defining their roles in the regulation of Purkinje neuron excitability. Here, we review this important work and highlight the remaining questions about the molecular mechanisms controlling the expression and the functioning of Nav currents in Purkinje neurons. We also discuss the impact of the dynamic regulation of Nav currents on the functioning of individual Purkinje neurons and cerebellar circuits.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, Box 8086, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Jeanne M Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Medicine, Washington University School of Medicine, Box 8086, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
| |
Collapse
|
32
|
Pappalardo LW, Samad OA, Liu S, Zwinger PJ, Black JA, Waxman SG. Nav1.5 in astrocytes plays a sex-specific role in clinical outcomes in a mouse model of multiple sclerosis. Glia 2018; 66:2174-2187. [PMID: 30194875 DOI: 10.1002/glia.23470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022]
Abstract
Astrogliosis is a hallmark of neuroinflammatory disorders such as multiple sclerosis (MS). A detailed understanding of the underlying molecular mechanisms governing astrogliosis might facilitate the development of therapeutic targets. We investigated whether Nav1.5 expression in astrocytes plays a role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine model of MS. We created a conditional knockout of Nav1.5 in astrocytes and determined whether this affects the clinical course of EAE, focal macrophage and T cell infiltration, and diffuse activation of astrocytes. We show that deletion of Nav1.5 from astrocytes leads to significantly worsened clinical outcomes in EAE, with increased inflammatory infiltrate in both early and late stages of disease, unexpectedly, in a sex-specific manner. Removal of Nav1.5 in astrocytes leads to increased inflammation in female mice with EAE, including increased astroglial response and infiltration of T cells and phagocytic monocytes. These cellular changes are consistent with more severe EAE clinical scores. Additionally, we found evidence suggesting possible dysregulation of the immune response-particularly with regard to infiltrating macrophages and activated microglia-in female Nav1.5 KO mice compared with WT littermate controls. Together, our results show that deletion of Nav1.5 from astrocytes leads to significantly worsened clinical outcomes in EAE, with increased inflammatory infiltrate in both early and late stages of disease, in a sex-specific manner.
Collapse
Affiliation(s)
- Laura W Pappalardo
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, 06516
| | - Omar A Samad
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, 06516
| | - Shujun Liu
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, 06516
| | - Pamela J Zwinger
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, 06516
| | - Joel A Black
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, 06516
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, 06510.,Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, 06516
| |
Collapse
|
33
|
The invasiveness of human cervical cancer associated to the function of Na V1.6 channels is mediated by MMP-2 activity. Sci Rep 2018; 8:12995. [PMID: 30158710 PMCID: PMC6115389 DOI: 10.1038/s41598-018-31364-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/16/2018] [Indexed: 12/17/2022] Open
Abstract
Voltage-gated sodium (NaV) channels have been related with cell migration and invasiveness in human cancers. We previously reported the contribution of NaV1.6 channels activity with the invasion capacity of cervical cancer (CeCa) positive to Human Papilloma Virus type 16 (HPV16), which accounts for 50% of all CeCa cases. Here, we show that NaV1.6 gene (SCN8A) overexpression is a general characteristic of CeCa, regardless of the HPV type. In contrast, no differences were observed in NaV1.6 channel expression between samples of non-cancerous and cervical intraepithelial neoplasia. Additionally, we found that CeCa cell lines, C33A, SiHa, CaSki and HeLa, express mainly the splice variant of SCN8A that lacks exon 18, shown to encode for an intracellularly localized NaV1.6 channel, whereas the full-length adult form was present in CeCa biopsies. Correlatively, patch-clamp experiments showed no evidence of whole-cell sodium currents (INa) in CeCa cell lines. Heterologous expression of full-length NaV1.6 isoform in C33A cells produced INa, which were sufficient to significantly increase invasion capacity and matrix metalloproteinase type 2 (MMP-2) activity. These data suggest that upregulation of NaV1.6 channel expression occurs when cervical epithelium have been transformed into cancer cells, and that NaV1.6-mediated invasiveness of CeCa cells involves MMP-2 activity. Thus, our findings support the notion about using NaV channels as therapeutic targets against cancer metastasis.
Collapse
|
34
|
Wagnon JL, Mencacci NE, Barker BS, Wengert ER, Bhatia KP, Balint B, Carecchio M, Wood NW, Patel MK, Meisler MH. Partial loss-of-function of sodium channel SCN8A in familial isolated myoclonus. Hum Mutat 2018; 39:965-969. [PMID: 29726066 PMCID: PMC6032947 DOI: 10.1002/humu.23547] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 02/01/2023]
Abstract
Variants in the neuronal sodium channel gene SCN8A have been implicated in several neurological disorders. Early infantile epileptic encephalopathy type 13 results from de novo gain‐of‐function mutations that alter the biophysical properties of the channel. Complete loss‐of‐function variants of SCN8A have been identified in cases of isolated intellectual disability. We now report a novel heterozygous SCN8A variant, p.Pro1719Arg, in a small pedigree with five family members affected with autosomal dominant upper limb isolated myoclonus without seizures or cognitive impairment. Functional analysis of the p.Pro1719Arg variant in transfected neuron‐derived cells demonstrated greatly reduced Nav1.6 channel activity without altered gating properties. Hypomorphic alleles of Scn8a in the mouse are known to result in similar movement disorders. This study expands the phenotypic and functional spectrum of SCN8A variants to include inherited nonepileptic isolated myoclonus. SCN8A can be considered as a candidate gene for isolated movement disorders without seizures.
Collapse
Affiliation(s)
- Jacy L Wagnon
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan
| | - Niccolò E Mencacci
- Department of Neurology, Northwestern University, Chicago, Illinois.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Bryan S Barker
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia.,Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Eric R Wengert
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia.,Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Kailash P Bhatia
- Sobell Department, Institute of Neurology, University College of London, London, UK
| | - Bettina Balint
- Sobell Department, Institute of Neurology, University College of London, London, UK
| | - Miryam Carecchio
- Molecular Neurogenetics Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy.,Department of Pediatric Neurology, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy.,Department of Medicine and Surgery, PhD Programme in Molecular and Translational Medicine, Milan Bicocca University, Monza, Italy
| | - Nicholas W Wood
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Manoj K Patel
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia.,Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan.,Department of Neurology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
35
|
Hoxha E, Balbo I, Miniaci MC, Tempia F. Purkinje Cell Signaling Deficits in Animal Models of Ataxia. Front Synaptic Neurosci 2018; 10:6. [PMID: 29760657 PMCID: PMC5937225 DOI: 10.3389/fnsyn.2018.00006] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022] Open
Abstract
Purkinje cell (PC) dysfunction or degeneration is the most frequent finding in animal models with ataxic symptoms. Mutations affecting intrinsic membrane properties can lead to ataxia by altering the firing rate of PCs or their firing pattern. However, the relationship between specific firing alterations and motor symptoms is not yet clear, and in some cases PC dysfunction precedes the onset of ataxic signs. Moreover, a great variety of ionic and synaptic mechanisms can affect PC signaling, resulting in different features of motor dysfunction. Mutations affecting Na+ channels (NaV1.1, NaV1.6, NaVβ4, Fgf14 or Rer1) reduce the firing rate of PCs, mainly via an impairment of the Na+ resurgent current. Mutations that reduce Kv3 currents limit the firing rate frequency range. Mutations of Kv1 channels act mainly on inhibitory interneurons, generating excessive GABAergic signaling onto PCs, resulting in episodic ataxia. Kv4.3 mutations are responsible for a complex syndrome with several neurologic dysfunctions including ataxia. Mutations of either Cav or BK channels have similar consequences, consisting in a disruption of the firing pattern of PCs, with loss of precision, leading to ataxia. Another category of pathogenic mechanisms of ataxia regards alterations of synaptic signals arriving at the PC. At the parallel fiber (PF)-PC synapse, mutations of glutamate delta-2 (GluD2) or its ligand Crbl1 are responsible for the loss of synaptic contacts, abolishment of long-term depression (LTD) and motor deficits. At the same synapse, a correct function of metabotropic glutamate receptor 1 (mGlu1) receptors is necessary to avoid ataxia. Failure of climbing fiber (CF) maturation and establishment of PC mono-innervation occurs in a great number of mutant mice, including mGlu1 and its transduction pathway, GluD2, semaphorins and their receptors. All these models have in common the alteration of PC output signals, due to a variety of mechanisms affecting incoming synaptic signals or the way they are processed by the repertoire of ionic channels responsible for intrinsic membrane properties. Although the PC is a final common pathway of ataxia, the link between specific firing alterations and neurologic symptoms has not yet been systematically studied and the alterations of the cerebellar contribution to motor signals are still unknown.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Ilaria Balbo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy.,National Institute of Neuroscience (INN), Turin, Italy
| |
Collapse
|
36
|
Sarnaik R, Raman IM. Control of voluntary and optogenetically perturbed locomotion by spike rate and timing of neurons of the mouse cerebellar nuclei. eLife 2018; 7:29546. [PMID: 29659351 PMCID: PMC5902160 DOI: 10.7554/elife.29546] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 03/30/2018] [Indexed: 11/13/2022] Open
Abstract
Neurons of the cerebellar nuclei (CbN), which generate cerebellar output, are inhibited by Purkinje cells. With extracellular recordings during voluntary locomotion in head-fixed mice, we tested how the rate and coherence of inhibition influence CbN cell firing and well-practiced movements. Firing rates of Purkinje and CbN cells were modulated systematically through the stride cycle (~200–300 ms). Optogenetically stimulating ChR2-expressing Purkinje cells with light steps or trains evoked either asynchronous or synchronous inhibition of CbN cells. Steps slowed CbN firing. Trains suppressed CbN cell firing less effectively, but consistently altered millisecond-scale spike timing. Steps or trains that perturbed stride-related modulation of CbN cell firing rates correlated well with irregularities of movement, suggesting that ongoing locomotion is sensitive to alterations in modulated CbN cell firing. Unperturbed locomotion continued more often during trains than steps, however, suggesting that stride-related modulation of CbN spiking is less readily disrupted by synchronous than asynchronous inhibition.
Collapse
Affiliation(s)
- Rashmi Sarnaik
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, United States
| |
Collapse
|
37
|
Aberrant Sodium Channel Currents and Hyperexcitability of Medial Entorhinal Cortex Neurons in a Mouse Model of SCN8A Encephalopathy. J Neurosci 2017; 37:7643-7655. [PMID: 28676574 DOI: 10.1523/jneurosci.2709-16.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 06/06/2017] [Accepted: 06/27/2017] [Indexed: 02/02/2023] Open
Abstract
SCN8A encephalopathy, or early infantile epileptic encephalopathy 13 (EIEE13), is caused predominantly by de novo gain-of-function mutations in the voltage-gated Na channel Nav1.6. Affected individuals suffer from refractory seizures, developmental delay, cognitive disability, and elevated risk of sudden unexpected death in epilepsy (SUDEP). A knock-in mouse model carrying the patient mutation p.Asn1768Asp (N1768D) reproduces many features of the disorder, including spontaneous seizures and SUDEP. We used the mouse model to examine the effects of the mutation on layer II stellate neurons of the medial entorhinal cortex (mEC), which transmit excitatory input to the hippocampus. Heterozygous (Scn8aD/+), homozygous (Scn8aD/D)), and WT (Scn8a+/+) littermates were compared at 3 weeks of age, the time of seizure onset for homozygous mice. Heterozygotes remain seizure free for another month. mEC layer II neurons of heterozygous and homozygous mice were hyperexcitable and generated long-lasting depolarizing potentials with bursts of action potentials after synaptic stimulation. Recording of Na currents revealed proexcitatory increases in persistent and resurgent currents and rightward shifts in inactivation parameters, leading to significant increases in the magnitude of window currents. The proexcitatory changes were more pronounced in homozygous mice than in heterozygotes, consistent with the earlier age of seizure onset in homozygotes. These studies demonstrate that the N1768D mutation increases the excitability of mEC layer II neurons by increasing persistent and resurgent Na currents and disrupting channel inactivation. The aberrant activities of mEC layer II neurons would provide excessive excitatory input to the hippocampus and contribute to hyperexcitability of hippocampal neurons in this model of SCN8A encephalopathy.SIGNIFICANCE STATEMENTSCN8A encephalopathy is a devastating neurological disorder that results from de novo mutations in the Na channel Nav1.6. In addition to seizures, patients suffer from cognitive and developmental delays and are at high risk for sudden unexpected death in epilepsy (SUDEP). A mouse knock-in model expressing the patient mutation N1768D reproduces several pathological phenotypes, including spontaneous seizures and sudden death. We demonstrate that medial entorhinal cortex (mEC) neurons from the mouse model exhibit proexcitatory alterations in Na channel activity, some of which were not seen in hippocampal or cortical neurons, and resulting in neuronal hyperexcitability. Because mEC neurons regulate the activity of the hippocampus, which plays an important role in seizure onset, we propose that these profound changes in mEC neuron excitability associated with the gain-of-function mutation of Nav1.6 may increase excitatory drive into the hippocampus, culminating in seizure activity and SUDEP.
Collapse
|
38
|
Wagnon JL, Barker BS, Ottolini M, Park Y, Volkheimer A, Valdez P, Swinkels MEM, Patel MK, Meisler MH. Loss-of-function variants of SCN8A in intellectual disability without seizures. NEUROLOGY-GENETICS 2017; 3:e170. [PMID: 28702509 PMCID: PMC5499976 DOI: 10.1212/nxg.0000000000000170] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/11/2017] [Indexed: 11/15/2022]
Abstract
Objective: To determine the functional effect of SCN8A missense mutations in 2 children with intellectual disability and developmental delay but no seizures. Methods: Genomic DNA was analyzed by next-generation sequencing. SCN8A variants were introduced into the Nav1.6 complementary DNA by site-directed mutagenesis. Channel activity was measured electrophysiologically in transfected ND7/23 cells. The stability of the mutant channels was assessed by Western blot. Results: Both children were heterozygous for novel missense variants that altered conserved residues in transmembrane segments of Nav1.6, p.Gly964Arg in D2S6 and p.Glu1218Lys in D3S1. Both altered amino acids are evolutionarily conserved in vertebrate and invertebrate channels and are predicted to be deleterious. Neither was observed in the general population. Both variants completely prevented the generation of sodium currents in transfected cells. The abundance of Nav1.6 protein was reduced by the Glu1218Lys substitution. Conclusions: Haploinsufficiency of SCN8A is associated with cognitive impairment. These observations extend the phenotypic spectrum of SCN8A mutations beyond their established role in epileptic encephalopathy (OMIM#614558) and other seizure disorders. SCN8A should be considered as a candidate gene for intellectual disability, regardless of seizure status.
Collapse
Affiliation(s)
- Jacy L Wagnon
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Bryan S Barker
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Matteo Ottolini
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Young Park
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Alicia Volkheimer
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Purnima Valdez
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Marielle E M Swinkels
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Manoj K Patel
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| | - Miriam H Meisler
- Department of Human Genetics (J.L.W., Y.P., M.H.M.), University of Michigan, Ann Arbor; Department of Anesthesiology (B.S.B., M.O., M.K.P.) and Neuroscience Graduate Program (B.S.B., M.K.P.), University of Virginia, Charlottesville; Department of Medicine (A.V.), Veterans Affairs Medical Center (A.V.), and Department of Pediatrics (P.V.), Duke University, Durham, NC; and Department of Medical Genetics (M.E.M.S.), University Medical Centre Utrecht, the Netherlands
| |
Collapse
|
39
|
Ransdell JL, Dranoff E, Lau B, Lo WL, Donermeyer DL, Allen PM, Nerbonne JM. Loss of Navβ4-Mediated Regulation of Sodium Currents in Adult Purkinje Neurons Disrupts Firing and Impairs Motor Coordination and Balance. Cell Rep 2017; 19:532-544. [PMID: 28423317 PMCID: PMC5473293 DOI: 10.1016/j.celrep.2017.03.068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/16/2017] [Accepted: 03/22/2017] [Indexed: 11/28/2022] Open
Abstract
The resurgent component of voltage-gated Na+ (Nav) currents, INaR, has been suggested to provide the depolarizing drive for high-frequency firing and to be generated by voltage-dependent Nav channel block (at depolarized potentials) and unblock (at hyperpolarized potentials) by the accessory Navβ4 subunit. To test these hypotheses, we examined the effects of the targeted deletion of Scn4b (Navβ4) on INaR and on repetitive firing in cerebellar Purkinje neurons. We show here that Scn4b-/- animals have deficits in motor coordination and balance and that firing rates in Scn4b-/- Purkinje neurons are markedly attenuated. Acute, in vivo short hairpin RNA (shRNA)-mediated "knockdown" of Navβ4 in adult Purkinje neurons also reduced spontaneous and evoked firing rates. Dynamic clamp-mediated addition of INaR partially rescued firing in Scn4b-/- Purkinje neurons. Voltage-clamp experiments revealed that INaR was reduced (by ∼50%), but not eliminated, in Scn4b-/- Purkinje neurons, revealing that additional mechanisms contribute to generation of INaR.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Departments of Developmental Biology and Internal Medicine , Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Edward Dranoff
- Departments of Developmental Biology and Internal Medicine , Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brandon Lau
- Departments of Developmental Biology and Internal Medicine , Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wan-Lin Lo
- Department of Pathology and Immunology , Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David L Donermeyer
- Department of Pathology and Immunology , Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul M Allen
- Department of Pathology and Immunology , Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeanne M Nerbonne
- Departments of Developmental Biology and Internal Medicine , Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
Valkova C, Liebmann L, Krämer A, Hübner CA, Kaether C. The sorting receptor Rer1 controls Purkinje cell function via voltage gated sodium channels. Sci Rep 2017; 7:41248. [PMID: 28117367 PMCID: PMC5259745 DOI: 10.1038/srep41248] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 12/19/2016] [Indexed: 01/08/2023] Open
Abstract
Rer1 is a sorting receptor in the early secretory pathway that controls the assembly and the cell surface transport of selected multimeric membrane protein complexes. Mice with a Purkinje cell (PC) specific deletion of Rer1 showed normal polarization and differentiation of PCs and normal development of the cerebellum. However, PC-specific loss of Rer1 led to age-dependent motor deficits in beam walk, ladder climbing and gait. Analysis of brain sections revealed a specific degeneration of PCs in the anterior cerebellar lobe in old animals. Electrophysiological recordings demonstrated severe deficits in spontaneous action potential generation. Measurements of resurgent currents indicated decreased surface densities of voltage-gated sodium channels (Nav), but not changes in individual channels. Analysis of mice with a whole brain Rer1-deletion demonstrated a strong down-regulation of Nav1.6 and 1.1 in the absence of Rer1, whereas protein levels of the related Cav2.1 and of Kv3.3 and 7.2 channels were not affected. The data suggest that Rer1 controls the assembly and transport of Nav1.1 and 1.6, the principal sodium channels responsible for recurrent firing, in PCs.
Collapse
Affiliation(s)
- Christina Valkova
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07743 Jena, Germany
| | - Lutz Liebmann
- Institut für Humangenetik, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Germany
| | - Andreas Krämer
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07743 Jena, Germany
| | - Christian A Hübner
- Institut für Humangenetik, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Germany
| | - Christoph Kaether
- Leibniz Institut für Alternsforschung-Fritz Lipmann Institut, 07743 Jena, Germany
| |
Collapse
|
41
|
Abstract
Alternative precursor-mRNA splicing is a key mechanism for regulating gene expression in mammals and is controlled by specialized RNA-binding proteins. The misregulation of splicing is implicated in multiple neurological disorders. We describe recent mouse genetic studies of alternative splicing that reveal its critical role in both neuronal development and the function of mature neurons. We discuss the challenges in understanding the extensive genetic programmes controlled by proteins that regulate splicing, both during development and in the adult brain.
Collapse
Affiliation(s)
- Celine K Vuong
- Molecular Biology Interdepartmental Graduate Program, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Douglas L Black
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Sika Zheng
- Division of Biomedical Sciences, University of California at Riverside, Riverside, California 92521, USA
| |
Collapse
|
42
|
Mercer AA, Palarz KJ, Tabatadze N, Woolley CS, Raman IM. Sex differences in cerebellar synaptic transmission and sex-specific responses to autism-linked Gabrb3 mutations in mice. eLife 2016; 5. [PMID: 27077953 PMCID: PMC4878876 DOI: 10.7554/elife.07596] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 04/13/2016] [Indexed: 12/18/2022] Open
Abstract
Neurons of the cerebellar nuclei (CbN) transmit cerebellar signals to premotor areas. The cerebellum expresses several autism-linked genes, including GABRB3, which encodes GABAA receptor β3 subunits and is among the maternal alleles deleted in Angelman syndrome. We tested how this Gabrb3 m-/p+ mutation affects CbN physiology in mice, separating responses of males and females. Wild-type mice showed sex differences in synaptic excitation, inhibition, and intrinsic properties. Relative to females, CbN cells of males had smaller synaptically evoked mGluR1/5-dependent currents, slower Purkinje-mediated IPSCs, and lower spontaneous firing rates, but rotarod performances were indistinguishable. In mutant CbN cells, IPSC kinetics were unchanged, but mutant males, unlike females, showed enlarged mGluR1/5 responses and accelerated spontaneous firing. These changes appear compensatory, since mutant males but not females performed indistinguishably from wild-type siblings on the rotarod task. Thus, sex differences in cerebellar physiology produce similar behavioral output, but provide distinct baselines for responses to mutations.
Collapse
Affiliation(s)
- Audrey A Mercer
- Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, United States.,Department of Neurobiology, Northwestern University, Evanston, United States
| | - Kristin J Palarz
- Department of Neurobiology, Northwestern University, Evanston, United States.,Integrated Science Program, Northwestern University, Evanston, United States
| | - Nino Tabatadze
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Catherine S Woolley
- Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, United States.,Department of Neurobiology, Northwestern University, Evanston, United States
| | - Indira M Raman
- Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, United States.,Department of Neurobiology, Northwestern University, Evanston, United States.,Integrated Science Program, Northwestern University, Evanston, United States
| |
Collapse
|
43
|
Handforth A. Linking Essential Tremor to the Cerebellum—Animal Model Evidence. THE CEREBELLUM 2015; 15:285-98. [DOI: 10.1007/s12311-015-0750-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
44
|
Blanchard MG, Willemsen MH, Walker JB, Dib-Hajj SD, Waxman SG, Jongmans MCJ, Kleefstra T, van de Warrenburg BP, Praamstra P, Nicolai J, Yntema HG, Bindels RJM, Meisler MH, Kamsteeg EJ. De novo gain-of-function and loss-of-function mutations of SCN8A in patients with intellectual disabilities and epilepsy. J Med Genet 2015; 52:330-7. [PMID: 25725044 PMCID: PMC4413743 DOI: 10.1136/jmedgenet-2014-102813] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/05/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Mutations of SCN8A encoding the neuronal voltage-gated sodium channel NaV1.6 are associated with early-infantile epileptic encephalopathy type 13 (EIEE13) and intellectual disability. Using clinical exome sequencing, we have detected three novel de novo SCN8A mutations in patients with intellectual disabilities, and variable clinical features including seizures in two patients. To determine the causality of these SCN8A mutations in the disease of those three patients, we aimed to study the (dys)function of the mutant sodium channels. METHODS The functional consequences of the three SCN8A mutations were assessed using electrophysiological analyses in transfected cells. Genotype-phenotype correlations of these and other cases were related to the functional analyses. RESULTS The first mutant displayed a 10 mV hyperpolarising shift in voltage dependence of activation (gain of function), the second did not form functional channels (loss of function), while the third mutation was functionally indistinguishable from the wildtype channel. CONCLUSIONS Comparison of the clinical features of these patients with those in the literature suggests that gain-of-function mutations are associated with severe EIEE, while heterozygous loss-of-function mutations cause intellectual disability with or without seizures. These data demonstrate that functional analysis of missense mutations detected by clinical exome sequencing, both inherited and de novo, is valuable for clinical interpretation in the age of massive parallel sequencing.
Collapse
Affiliation(s)
- Maxime G Blanchard
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjolein H Willemsen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jaclyn B Walker
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sulayman D Dib-Hajj
- The Center for Neuroscience & Regeneration Research, Yale School of Medicine, New Haven, Connecticut, USA The Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Stephen G Waxman
- The Center for Neuroscience & Regeneration Research, Yale School of Medicine, New Haven, Connecticut, USA The Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Marjolijn C J Jongmans
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tjitske Kleefstra
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bart P van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Praamstra
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost Nicolai
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands Epilepsy Center Kempenhaeghe, Heeze, The Netherlands
| | - Helger G Yntema
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
45
|
Saba L, Viscomi MT, Caioli S, Pignataro A, Bisicchia E, Pieri M, Molinari M, Ammassari-Teule M, Zona C. Altered Functionality, Morphology, and Vesicular Glutamate Transporter Expression of Cortical Motor Neurons from a Presymptomatic Mouse Model of Amyotrophic Lateral Sclerosis. Cereb Cortex 2015; 26:1512-28. [DOI: 10.1093/cercor/bhu317] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
46
|
Hess EJ, Jinnah H. Mouse Models of Dystonia. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00027-5] [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
|
47
|
Martin NJ, Ferreiro SF, Barbault F, Nicolas M, Lecellier G, Paetz C, Gaysinski M, Alonso E, Thomas OP, Botana LM, Raharivelomanana P. Indole alkaloids from the Marquesan plant Rauvolfia nukuhivensis and their effects on ion channels. PHYTOCHEMISTRY 2015; 109:84-95. [PMID: 25468537 DOI: 10.1016/j.phytochem.2014.10.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/12/2014] [Accepted: 10/20/2014] [Indexed: 06/04/2023]
Abstract
In addition to the already reported nukuhivensiums 1 and 2, 11 indole alkaloids were isolated from the bark of the plant Rauvolfia nukuhivensis, growing in the Marquesas archipelago. The known sandwicine (3), isosandwicine (4), spegatrine (8), lochneram (9), flavopereirine (13) have been found in this plant together with the norsandwicine (5), isonorsandwicine (6), Nb-methylisosandwicine (7), 10-methoxypanarine (10), nortueiaoine (11), tueiaoine (12). The structure elucidation was performed on the basis of a deep exploration of the NMR and HRESIMS data as well as comparison with literature data for similar compounds. Norsandwicine, 10-methoxypanarine, tueiaoine, and more importantly nukuhivensiums, were shown to significantly induce a reduction of IKr amplitude (HERG current). Molecular modelling through docking was performed in order to illustrate this result.
Collapse
Affiliation(s)
- Nicolas J Martin
- Laboratoire EIMS UMR 241 EIO, Université de la Polynésie française, BP 6570, 98702 Faa'a, Tahiti, French Polynesia.
| | - Sara F Ferreiro
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.
| | - Florent Barbault
- Laboratoire ITODYS UMR CNRS 7086, Université Paris Diderot, Sorbonne Paris Cité, 15 rue J.-A. de Baïf, 75013 Paris, France.
| | - Mael Nicolas
- Laboratoire EIMS UMR 241 EIO, Université de la Polynésie française, BP 6570, 98702 Faa'a, Tahiti, French Polynesia.
| | - Gaël Lecellier
- Université de Versailles Saint Quentin en Yvelines, 55 Avenue des Etats-Unis, 78035 Versailles Cedex, France; Laboratoire d'Excellence "CORAIL" - Centre de Recherches Insulaires et Observatoire de l'Environnement (CRIOBE), BP 1013 - 98729, Papetoai, Moorea, French Polynesia.
| | - Christian Paetz
- MPI for Chemical Ecology, Hans-Knöll-Strasse 8, 07745 Jena, Germany.
| | - Marc Gaysinski
- Institut de Chimie de Nice-PCRE, UMR 7272 CNRS, Université de Nice-Sophia Antipolis, Parc Valrose, 06108 Nice, France.
| | - Eva Alonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.
| | - Olivier P Thomas
- Institut de Chimie de Nice-PCRE, UMR 7272 CNRS, Université de Nice-Sophia Antipolis, Parc Valrose, 06108 Nice, France; Institut Méditerranéen de Biodiversité et d́Ecologie marine et continentale, UMR 7263 CNRS - IRD - Aix-Marseille Université - UAPV, Station Marine d́Endoume, Rue de la Batterie des Lions, 13007 Marseille, France.
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, Spain.
| | - Phila Raharivelomanana
- Laboratoire EIMS UMR 241 EIO, Université de la Polynésie française, BP 6570, 98702 Faa'a, Tahiti, French Polynesia.
| |
Collapse
|
48
|
Brunklaus A, Ellis R, Reavey E, Semsarian C, Zuberi SM. Genotype phenotype associations across the voltage-gated sodium channel family. J Med Genet 2014; 51:650-8. [PMID: 25163687 DOI: 10.1136/jmedgenet-2014-102608] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mutations in genes encoding voltage-gated sodium channels have emerged as the most clinically relevant genes associated with epilepsy, cardiac conduction defects, skeletal muscle channelopathies and peripheral pain disorders. Geneticists in partnership with neurologists and cardiologists are often asked to comment on the clinical significance of specific mutations. We have reviewed the evidence relating to genotype phenotype associations among the best known voltage-gated sodium channel related disorders. Comparing over 1300 sodium channel mutations in central and peripheral nervous system, heart and muscle, we have identified many similarities in the genetic and clinical characteristics across the voltage-gated sodium channel family. There is evidence, that the level of impairment a specific mutation causes can be anticipated by the underlying physico-chemical property change of that mutation. Across missense mutations those with higher Grantham scores are associated with more severe phenotypes and truncating mutations underlie the most severe phenotypes. Missense mutations are clustered in specific areas and are associated with distinct phenotypes according to their position in the protein. Inherited mutations tend to be less severe than de novo mutations which are usually associated with greater physico-chemical difference. These findings should lead to a better understanding of the clinical significance of specific voltage-gated sodium channel mutations, aiding geneticists and physicians in the interpretation of genetic variants and counselling individuals and their families.
Collapse
Affiliation(s)
- Andreas Brunklaus
- The Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow, UK
| | - Rachael Ellis
- The Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow, UK Molecular Diagnostics, West of Scotland Genetic Services, Southern General Hospital, Glasgow, UK
| | - Eleanor Reavey
- The Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow, UK Molecular Diagnostics, West of Scotland Genetic Services, Southern General Hospital, Glasgow, UK
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia Sydney Medical School, University of Sydney, Australia
| | - Sameer M Zuberi
- The Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow, UK School of Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, UK
| |
Collapse
|
49
|
Luna-Cancalon K, Sikora KM, Pappas SS, Singh V, Wulff H, Paulson HL, Burmeister M, Shakkottai VG. Alterations in cerebellar physiology are associated with a stiff-legged gait in Atcay(ji-hes) mice. Neurobiol Dis 2014; 67:140-8. [PMID: 24727095 DOI: 10.1016/j.nbd.2014.03.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/25/2014] [Accepted: 03/31/2014] [Indexed: 01/17/2023] Open
Abstract
Recent evidence suggests that dystonia, a movement disorder characterized by sustained involuntary muscle contractions, can be associated with cerebellar abnormalities. The basis for how functional changes in the cerebellum can cause dystonia is poorly understood. Here we identify alterations in physiology in Atcay(ji-hes) mice which in addition to ataxia, have an abnormal gait with hind limb extension and toe walking, reminiscent of human dystonic gait. No morphological abnormalities in the brain accompany the dystonia, but partial cerebellectomy causes resolution of the stiff-legged gait, suggesting that cerebellar dysfunction contributes to the dystonic gait of Atcay(ji-hes) mice. Recordings from Purkinje and deep cerebellar nuclear (DCN) neurons in acute brain slices were used to determine the physiological correlates of dystonia in the Atcay(ji-hes) mice. Approximately 50% of cerebellar Purkinje neurons fail to display the normal repetitive firing characteristic of these cells. In addition, DCN neurons exhibit increased intrinsic firing frequencies with a subset of neurons displaying bursts of action potentials. This increased intrinsic excitability of DCN neurons is accompanied by a reduction in after-hyperpolarization currents mediated by small-conductance calcium-activated potassium (SK) channels. An activator of SK channels reduces DCN neuron firing frequency in acute cerebellar slices and improves the dystonic gait of Atcay(ji-hes) mice. These results suggest that a combination of reduced Purkinje neuron activity and increased DCN intrinsic excitability can result in a combination of ataxia and a dystonia-like gait in mice.
Collapse
Affiliation(s)
| | - Kristine M Sikora
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samuel S Pappas
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margit Burmeister
- Molecular & Behavioral Neuroscience Institute, Departments of Psychiatry, Computational Medicine & Bioinformatics and Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
50
|
Feng SS, Lin R, Gauck V, Jaeger D. Gain control of synaptic response function in cerebellar nuclear neurons by a calcium-activated potassium conductance. THE CEREBELLUM 2014; 12:692-706. [PMID: 23605187 DOI: 10.1007/s12311-013-0476-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Small conductance Ca(2+)-activated potassium (SK) current provides an important modulator of excitatory synaptic transmission, which undergoes plastic regulation via multiple mechanisms. We examined whether inhibitory input processing is also dependent on SK current in the cerebellar nuclei (CN) where inhibition provides the only route of information transfer from the cerebellar cortical Purkinje cells. We employed dynamic clamping in conjunction with computer simulations to address this question. We found that SK current plays a critical role in the inhibitory synaptic control of spiking output. Specifically, regulation of SK current density resulted in a gain control of spiking output, such that low SK current promoted large output signaling for large inhibitory cell input fluctuations due to Purkinje cell synchronization. In contrast, smaller nonsynchronized Purkinje cell input fluctuations were not amplified. Regulation of SK density in the CN therefore would likely lead to important consequences for the transmission of synchronized Purkinje cell activity to the motor system.
Collapse
Affiliation(s)
- Steven Si Feng
- Department of Biology, Emory University, 1510 Clifton Rd., Atlanta, GA, 30322, USA
| | | | | | | |
Collapse
|