1
|
Lauerer RJ, Lerche H. Voltage-gated calcium channels in genetic epilepsies. J Neurochem 2024; 168:3853-3871. [PMID: 37822150 PMCID: PMC11591408 DOI: 10.1111/jnc.15983] [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: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Voltage-gated calcium channels (VGCC) are abundant in the central nervous system and serve a broad spectrum of functions, either directly in cellular excitability or indirectly to regulate Ca2+ homeostasis. Ca2+ ions act as one of the main connections in excitation-transcription coupling, muscle contraction and excitation-exocytosis coupling, including synaptic transmission. In recent years, many genes encoding VGCCs main α or additional auxiliary subunits have been associated with epilepsy. This review sums up the current state of knowledge on disease mechanisms and provides guidance on disease-specific therapies where applicable.
Collapse
Affiliation(s)
- Robert J. Lauerer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| |
Collapse
|
2
|
Leitch B. Molecular Mechanisms Underlying the Generation of Absence Seizures: Identification of Potential Targets for Therapeutic Intervention. Int J Mol Sci 2024; 25:9821. [PMID: 39337309 PMCID: PMC11432152 DOI: 10.3390/ijms25189821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Understanding the molecular mechanisms underlying the generation of absence seizures is crucial for developing effective, patient-specific treatments for childhood absence epilepsy (CAE). Currently, one-third of patients remain refractive to the antiseizure medications (ASMs), previously called antiepileptic drugs (AEDs), available to treat CAE. Additionally, these ASMs often produce serious side effects and can even exacerbate symptoms in some patients. Determining the precise cellular and molecular mechanisms directly responsible for causing this type of epilepsy has proven challenging as they appear to be complex and multifactorial in patients with different genetic backgrounds. Aberrant neuronal activity in CAE may be caused by several mechanisms that are not fully understood. Thus, dissecting the causal factors that could be targeted in the development of precision medicines without side effects remains a high priority and the ultimate goal in this field of epilepsy research. The aim of this review is to highlight our current understanding of potential causative mechanisms for absence seizure generation, based on the latest research using cutting-edge technologies. This information will be important for identifying potential targets for future therapeutic intervention.
Collapse
Affiliation(s)
- Beulah Leitch
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
3
|
Soeung V, Puchalski RB, Noebels JL. The complex molecular epileptogenesis landscape of glioblastoma. Cell Rep Med 2024; 5:101691. [PMID: 39168100 PMCID: PMC11384957 DOI: 10.1016/j.xcrm.2024.101691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/30/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
The cortical microenvironment surrounding malignant glioblastoma is a source of depolarizing crosstalk favoring hyperexcitability, tumor expansion, and immune evasion. Neosynaptogenesis, excess glutamate, and altered intrinsic membrane currents contribute to excitability dyshomeostasis, yet only half of the cases develop seizures, suggesting that tumor and host genomics, along with location, rather than mass effect, play a critical role. We analyzed the spatial contours and expression of 358 clinically validated human epilepsy genes in the human glioblastoma transcriptome compared to non-tumor adult and developing cortex datasets. Nearly half, including dosage-sensitive genes whose expression levels are securely linked to monogenic epilepsy, are strikingly enriched and aberrantly regulated at the leading edge, supporting a complex epistatic basis for peritumoral epileptogenesis. Surround hyperexcitability induced by complex patterns of proepileptic gene expression may explain the limited efficacy of narrowly targeted antiseizure medicines and the persistence of epilepsy following tumor resection and clarify why not all brain tumors provoke seizures.
Collapse
Affiliation(s)
- Victoria Soeung
- Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Ralph B Puchalski
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Jeffrey L Noebels
- Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX, USA; Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Lindquist BE, Timbie C, Voskobiynyk Y, Paz JT. Thalamocortical circuits in generalized epilepsy: Pathophysiologic mechanisms and therapeutic targets. Neurobiol Dis 2023; 181:106094. [PMID: 36990364 PMCID: PMC10192143 DOI: 10.1016/j.nbd.2023.106094] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/02/2023] [Accepted: 03/19/2023] [Indexed: 03/29/2023] Open
Abstract
Generalized epilepsy affects 24 million people globally; at least 25% of cases remain medically refractory. The thalamus, with widespread connections throughout the brain, plays a critical role in generalized epilepsy. The intrinsic properties of thalamic neurons and the synaptic connections between populations of neurons in the nucleus reticularis thalami and thalamocortical relay nuclei help generate different firing patterns that influence brain states. In particular, transitions from tonic firing to highly synchronized burst firing mode in thalamic neurons can cause seizures that rapidly generalize and cause altered awareness and unconsciousness. Here, we review the most recent advances in our understanding of how thalamic activity is regulated and discuss the gaps in our understanding of the mechanisms of generalized epilepsy syndromes. Elucidating the role of the thalamus in generalized epilepsy syndromes may lead to new opportunities to better treat pharmaco-resistant generalized epilepsy by thalamic modulation and dietary therapy.
Collapse
Affiliation(s)
- Britta E Lindquist
- UCSF Department of Neurology, Division of Neurocritical Care, United States of America; UCSF Department of Neurology, Division of Pediatric Epilepsy, United States of America; UCSF Department of Neurology, United States of America
| | - Clare Timbie
- Gladstone Institute of Neurological Disease, United States of America; UCSF Department of Neurology, Division of Pediatric Epilepsy, United States of America; UCSF Department of Neurology, United States of America
| | - Yuliya Voskobiynyk
- Gladstone Institute of Neurological Disease, United States of America; UCSF Department of Neurology, United States of America
| | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, United States of America; UCSF Department of Neurology, United States of America; Kavli Institute for Fundamental Neuroscience, UCSF, United States of America.
| |
Collapse
|
5
|
Pulvirenti G, Caccamo M, Lo Bianco M, Mazzurco M, Praticò ER, Giallongo A, Gangi G, Zanghì A, Falsaperla R. Calcium Channels Genes and Their Epilepsy Phenotypes. JOURNAL OF PEDIATRIC NEUROLOGY 2023; 21:224-234. [DOI: 10.1055/s-0041-1728684] [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
AbstractCalcium (Ca2+) channel gene mutations play an important role in the pathogenesis of neurological episodic disorders like epilepsy. CACNA1A and CACNA1H genes are involved in the synthesis of calcium channels. Mutations in the α1A subunit of the P/Q type voltage-gated calcium channel gene (CACNA1A) located in 19p13.13, which encodes for the transmembrane pore-forming subunit of CAV2.1 voltage-dependent calcium channel, have been correlated to a large clinical spectrum of epilepsy such as idiopathic genetic epilepsy, early infantile epilepsy, and febrile seizures. Moreover, CACNA1A mutations have been demonstrated to be involved in spinocerebellar ataxia type 6, familiar hemiplegic migraine, episodic ataxia type 2, early-onset encephalopathy, and hemiconvulsion–hemiplegia epilepsy syndrome. This wide phenotype heterogeneity associated with CACNA1A mutations is correlated to different clinical and electrophysiological manifestations. CACNA1H gene, located in 16p13.3, encodes the α1H subunit of T-type calcium channel, expressing the transmembrane pore-forming subunit Cav3.2. Despite data still remain controversial, it has been identified as an important gene whose mutations seem strictly related to the pathogenesis of childhood absence epilepsy and other generalized epilepsies. The studied variants are mainly gain-of-function, hence responsible for an increase in neuronal susceptibility to seizures. CACNA1H mutations have also been associated with autism spectrum disorder and other behavior disorders. More recently, also amyotrophic lateral sclerosis has been related to CACNA1H alterations. The aim of this review, other than describe the CACNA1A and CACNA1H gene functions, is to identify mutations reported in literature and to analyze their possible correlations with specific epileptic disorders, purposing to guide an appropriate medical treatment recommendation.
Collapse
Affiliation(s)
- Giulio Pulvirenti
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Martina Caccamo
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Manuela Lo Bianco
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | | | - Alessandro Giallongo
- 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
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| |
Collapse
|
6
|
Gray MM, Naik A, Ebner TJ, Carter RE. Altered brain state during episodic dystonia in tottering mice decouples primary motor cortex from limb kinematics. DYSTONIA 2023; 2:10974. [PMID: 37800168 PMCID: PMC10554815 DOI: 10.3389/dyst.2023.10974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Episodic Ataxia Type 2 (EA2) is a rare neurological disorder caused by a mutation in the CACNA1A gene, encoding the P/Q-type voltage-gated Ca2+ channel important for neurotransmitter release. Patients with this channelopathy exhibit both cerebellar and cerebral pathologies, suggesting the condition affects both regions. The tottering (tg/tg) mouse is the most commonly used EA2 model due to an orthologous mutation in the cacna1a gene. The tg/tg mouse has three prominent behavioral phenotypes: a dramatic episodic dystonia; absence seizures with generalized spike and wave discharges (GSWDs); and mild ataxia. We previously observed a novel brain state, transient low-frequency oscillations (LFOs) in the cerebellum and cerebral cortex under anesthesia. In this study, we examine the relationships among the dystonic attack, GSWDs, and LFOs in the cerebral cortex. Previous studies characterized LFOs in the motor cortex of anesthetized tg/tg mice using flavoprotein autofluorescence imaging testing the hypothesis that LFOs provide a mechanism for the paroxysmal dystonia. We sought to obtain a more direct understanding of motor cortex (M1) activity during the dystonic episodes. Using two-photon Ca2+ imaging to investigate neuronal activity in M1 before, during, and after the dystonic attack, we show that there is not a significant change in the activity of M1 neurons from baseline through the attack. We also conducted simultaneous, multi-electrode recordings to further understand how M1 cellular activity and local field potentials change throughout the progression of the dystonic attack. Neither putative pyramidal nor inhibitory interneuron firing rate changed during the dystonic attack. However, we did observe a near complete loss of GSWDs during the dystonic attack in M1. Finally, using spike triggered averaging to align simultaneously recorded limb kinematics to the peak Ca2+ response, and vice versa, revealed a reduction in the spike triggered average during the dystonic episodes. Both the loss of GSWDs and the reduction in the coupling suggest that, during the dystonic attack, M1 is effectively decoupled from other structures. Overall, these results indicate that the attack is not initiated or controlled in M1, but elsewhere in the motor circuitry. The findings also highlight that LFOs, GSWDs, and dystonic attacks represent three brain states in tg/tg mice.
Collapse
Affiliation(s)
- Madelyn M Gray
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Anant Naik
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Timothy J Ebner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Russell E Carter
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
7
|
Amadori E, Pellino G, Bansal L, Mazzone S, Møller RS, Rubboli G, Striano P, Russo A. Answer to: Genetic paroxysmal neurological disorders featuring episodic ataxia and epilepsy (Amadori E et al., 2022). EJMG-D-22-00384. Eur J Med Genet 2022; 65:104634. [PMID: 36198373 DOI: 10.1016/j.ejmg.2022.104634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 09/28/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Elisabetta Amadori
- IRCCS G. Gaslini Institute, 16147, Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Italy
| | - Giuditta Pellino
- Pediatric Unit, Azienda USL Ferrara - Sant'Anna University Hospital of Ferrara, Ferrara, Italy; Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Lalit Bansal
- Division of Neurology, Children's Mercy Hospital, University of Missouri-Kansas City, Missouri, United States
| | - Serena Mazzone
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria dell'età pediatrica, Bologna, Italy
| | - Rikke S Møller
- Department for Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark; Institute for Regional Health Services Research, University of Southern Denmark, Odense, Denmark
| | - Guido Rubboli
- Department of Neurology, Danish Epilepsy Center (Member of the European Reference Network EpiCARE), Dianalund, University of Copenhagen, Copenhagen, Denmark
| | - Pasquale Striano
- IRCCS G. Gaslini Institute, 16147, Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Italy
| | - Angelo Russo
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria dell'età pediatrica, Bologna, Italy.
| |
Collapse
|
8
|
Schwitalla JC, Pakusch J, Mücher B, Brückner A, Depke DA, Fenzl T, De Zeeuw CI, Kros L, Hoebeek FE, Mark MD. Controlling absence seizures from the cerebellar nuclei via activation of the G q signaling pathway. Cell Mol Life Sci 2022; 79:197. [PMID: 35305155 PMCID: PMC8934336 DOI: 10.1007/s00018-022-04221-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/27/2022] [Accepted: 02/23/2022] [Indexed: 11/28/2022]
Abstract
Absence seizures (ASs) are characterized by pathological electrographic oscillations in the cerebral cortex and thalamus, which are called spike-and-wave discharges (SWDs). Subcortical structures, such as the cerebellum, may well contribute to the emergence of ASs, but the cellular and molecular underpinnings remain poorly understood. Here we show that the genetic ablation of P/Q-type calcium channels in cerebellar granule cells (quirky) or Purkinje cells (purky) leads to recurrent SWDs with the purky model showing the more severe phenotype. The quirky mouse model showed irregular action potential firing of their cerebellar nuclei (CN) neurons as well as rhythmic firing during the wave of their SWDs. The purky model also showed irregular CN firing, in addition to a reduced firing rate and rhythmicity during the spike of the SWDs. In both models, the incidence of SWDs could be decreased by increasing CN activity via activation of the Gq-coupled designer receptor exclusively activated by designer drugs (DREADDs) or via that of the Gq-coupled metabotropic glutamate receptor 1. In contrast, the incidence of SWDs was increased by decreasing CN activity via activation of the inhibitory Gi/o-coupled DREADD. Finally, disrupting CN rhythmic firing with a closed-loop channelrhodopsin-2 stimulation protocol confirmed that ongoing SWDs can be ceased by activating CN neurons. Together, our data highlight that P/Q-type calcium channels in cerebellar granule cells and Purkinje cells can be relevant for epileptogenesis, that Gq-coupled activation of CN neurons can exert anti-epileptic effects and that precisely timed activation of the CN can be used to stop ongoing SWDs.
Collapse
Affiliation(s)
| | - Johanna Pakusch
- Department of Behavioral Neuroscience, Ruhr-University Bochum, 44801, Bochum, Germany
| | - Brix Mücher
- Department of Zoology and Neurobiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - Alexander Brückner
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Dominic Alexej Depke
- European Institute of Molecular Imaging, University of Münster, 48149, Münster, Germany
| | - Thomas Fenzl
- Department of Anesthesiology and Intensive Care, TUM School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, 3015 AA, Rotterdam, The Netherlands.,Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, 1105, BA, Amsterdam, The Netherlands
| | - Lieke Kros
- Department of Neuroscience, Erasmus MC, 3015 AA, Rotterdam, The Netherlands
| | - Freek E Hoebeek
- Department for Developmental Origins of Disease, Wilhelmina Children's Hospital and Brain Center, University Medical Center Utrecht, 3584 EA, Utrecht, The Netherlands
| | - Melanie D Mark
- Department of Behavioral Neuroscience, Ruhr-University Bochum, 44801, Bochum, Germany.
| |
Collapse
|
9
|
Coulter I, Timic Stamenic T, Eggan P, Fine BR, Corrigan T, Covey DF, Yang L, Pan JQ, Todorovic SM. Different roles of T-type calcium channel isoforms in hypnosis induced by an endogenous neurosteroid epipregnanolone. Neuropharmacology 2021; 197:108739. [PMID: 34339750 PMCID: PMC8478885 DOI: 10.1016/j.neuropharm.2021.108739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Many neuroactive steroids induce sedation/hypnosis by potentiating γ-aminobutyric acid (GABAA) currents. However, we previously demonstrated that an endogenous neuroactive steroid epipregnanolone [(3β,5β)-3-hydroxypregnan-20-one] (EpiP) exerts potent peripheral analgesia and blocks T-type calcium currents while sparing GABAA currents in rat sensory neurons. This study seeks to investigate the behavioral effects elicited by systemic administration of EpiP and to characterize its use as an adjuvant agent to commonly used general anesthetics (GAs). METHODS Here, we utilized electroencephalographic (EEG) recordings to characterize thalamocortical oscillations, as well as behavioral assessment and mouse genetics with wild-type (WT) and different knockout (KO) models of T-channel isoforms to investigate potential sedative/hypnotic and immobilizing properties of EpiP. RESULTS Consistent with increased oscillations in slower EEG frequencies, EpiP induced an hypnotic state in WT mice when injected alone intra-peritoneally (i.p.) and effectively facilitated anesthetic effects of isoflurane (ISO) and sevoflurane (SEVO). The CaV3.1 (Cacna1g) KO mice demonstrated decreased sensitivity to EpiP-induced hypnosis when compared to WT mice, whereas no significant difference was noted between CaV3.2 (Cacna1h), CaV3.3 (Cacna1i) and WT mice. Finally, when compared to WT mice, onset of EpiP-induced hypnosis was delayed in CaV3.2 KO mice but not in CaV3.1 and CaV3.3 KO mice. CONCLUSION We posit that EpiP may have an important role as novel hypnotic and/or adjuvant to volatile anesthetic agents. We speculate that distinct hypnotic effects of EpiP across all three T-channel isoforms is due to their differential expression in thalamocortical circuitry.
Collapse
Affiliation(s)
- Ian Coulter
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Pierce Eggan
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Brier R. Fine
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Timothy Corrigan
- Department of Pediatrics, Division of Neurology,
Translational Epilepsy Research Program, University of Colorado, Anschutz Medical
Campus, Aurora, CO 80045, USA
| | - Douglas F. Covey
- Department of Developmental Biology, Washington University
School of Medicine, St. Louis, MO 63110, USA;,Taylor Family Institute for Innovative Psychiatric
Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lingling Yang
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Slobodan M. Todorovic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045;,Neuroscience, University of Colorado, Anschutz Medical
Campus, Aurora 80045;,Pharmacology Graduate Programs, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| |
Collapse
|
10
|
Lou S, Cui S. Drug treatment of epilepsy: From serendipitous discovery to evolutionary mechanisms. Curr Med Chem 2021; 29:3366-3391. [PMID: 34514980 DOI: 10.2174/0929867328666210910124727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 11/22/2022]
Abstract
Epilepsy is a chronic brain disorder caused by abnormal firing of neurons. Up to now, using antiepileptic drugs is the main method of epilepsy treatment. The development of antiepileptic drugs lasted for centuries. In general, most agents entering clinical practice act on the balance mechanisms of brain "excitability-inhibition". More specifically, they target voltage-gated ion channels, GABAergic transmission and glutamatergic transmission. In recent years, some novel drugs representing new mechanisms of action have been discovered. Although there are about 30 available drugs in the market, it is still in urgent need of discovering more effective and safer drugs. The development of new antiepileptic drugs is into a new era: from serendipitous discovery to evolutionary mechanism-based design. This article presents an overview of drug treatment of epilepsy, including a series of traditional and novel drugs.
Collapse
Affiliation(s)
- Shengying Lou
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| |
Collapse
|
11
|
Yu D, Febbo IG, Maroteaux MJ, Wang H, Song Y, Han X, Sun C, Meyer EE, Rowe S, Chen Y, Canavier CC, Schrader LA. The Transcription Factor Shox2 Shapes Neuron Firing Properties and Suppresses Seizures by Regulation of Key Ion Channels in Thalamocortical Neurons. Cereb Cortex 2021; 31:3194-3212. [PMID: 33675359 PMCID: PMC8196244 DOI: 10.1093/cercor/bhaa414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 01/02/2023] Open
Abstract
Thalamocortical neurons (TCNs) play a critical role in the maintenance of thalamocortical oscillations, dysregulation of which can result in certain types of seizures. Precise control over firing rates of TCNs is foundational to these oscillations, yet the transcriptional mechanisms that constrain these firing rates remain elusive. We hypothesized that Shox2 is a transcriptional regulator of ion channels important for TCN function and that loss of Shox2 alters firing frequency and activity, ultimately perturbing thalamocortical oscillations into an epilepsy-prone state. In this study, we used RNA sequencing and quantitative PCR of control and Shox2 knockout mice to determine Shox2-affected genes and revealed a network of ion channel genes important for neuronal firing properties. Protein regulation was confirmed by Western blotting, and electrophysiological recordings showed that Shox2 KO impacted the firing properties of a subpopulation of TCNs. Computational modeling showed that disruption of these conductances in a manner similar to Shox2's effects modulated frequency of oscillations and could convert sleep spindles to near spike and wave activity, which are a hallmark for absence epilepsy. Finally, Shox2 KO mice were more susceptible to pilocarpine-induced seizures. Overall, these results reveal Shox2 as a transcription factor important for TCN function in adult mouse thalamus.
Collapse
Affiliation(s)
- Diankun Yu
- Neuroscience Program, Brain Institute, Tulane University, USA
| | | | | | - Hanyun Wang
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Yingnan Song
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Xiao Han
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Cheng Sun
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Emily E Meyer
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Stuart Rowe
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Yiping Chen
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Carmen C Canavier
- Cell Biology and Anatomy, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Laura A Schrader
- Neuroscience Program, Brain Institute, Tulane University, USA
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
12
|
Eelkman Rooda OHJ, Kros L, Faneyte SJ, Holland PJ, Gornati SV, Poelman HJ, Jansen NA, Tolner EA, van den Maagdenberg AMJM, De Zeeuw CI, Hoebeek FE. Single-pulse stimulation of cerebellar nuclei stops epileptic thalamic activity. Brain Stimul 2021; 14:861-872. [PMID: 34022430 DOI: 10.1016/j.brs.2021.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/05/2021] [Accepted: 05/03/2021] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Epileptic (absence) seizures in the cerebral cortex can be stopped by pharmacological and optogenetic stimulation of the cerebellar nuclei (CN) neurons that innervate the thalamus. However, it is unclear how such stimulation can modify underlying thalamo-cortical oscillations. HYPOTHESIS Here we tested whether rhythmic synchronized thalamo-cortical activity during absence seizures can be desynchronized by single-pulse optogenetic stimulation of CN neurons to stop seizure activity. METHODS We performed simultaneous thalamic single-cell and electrocorticographical recordings in awake tottering mice, a genetic model of absence epilepsy, to investigate the rhythmicity and synchronicity. Furthermore, we tested interictally the impact of single-pulse optogenetic CN stimulation on thalamic and cortical recordings. RESULTS We show that thalamic firing is highly rhythmic and synchronized with cortical spike-and-wave discharges during absence seizures and that this phase-locked activity can be desynchronized upon single-pulse optogenetic stimulation of CN neurons. Notably, this stimulation of CN neurons was more effective in stopping seizures than direct, focal stimulation of groups of afferents innervating the thalamus. During interictal periods, CN stimulation evoked reliable but heterogeneous responses in thalamic cells in that they could show an increase or decrease in firing rate at various latencies, bi-phasic responses with an initial excitatory and subsequent inhibitory response, or no response at all. CONCLUSION Our data indicate that stimulation of CN neurons and their fibers in thalamus evokes differential effects in its downstream pathways and desynchronizes phase-locked thalamic neuronal firing during seizures, revealing a neurobiological mechanism that may explain how cerebellar stimulation can stop seizures.
Collapse
Affiliation(s)
- Oscar H J Eelkman Rooda
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands; Department of Neurosurgery, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands
| | - Lieke Kros
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands
| | - Sade J Faneyte
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands
| | - Peter J Holland
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Simona V Gornati
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands
| | - Huub J Poelman
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands
| | - Nico A Jansen
- Department of Neurology, Leiden University Medical Center, 2300, RC Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, 2300, RC Leiden, the Netherlands
| | - Else A Tolner
- Department of Neurology, Leiden University Medical Center, 2300, RC Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, 2300, RC Leiden, the Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Center, 2300, RC Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, 2300, RC Leiden, the Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands; Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, 1105, BA Amsterdam, the Netherlands
| | - Freek E Hoebeek
- Department of Neuroscience, Erasmus Medical Center, 3015, AA Rotterdam, the Netherlands; Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht Medical Center, 3508, AB Utrecht, the Netherlands.
| |
Collapse
|
13
|
Verriello L, Pauletto G, Nilo A, Lonigro I, Betto E, Valente M, Curcio F, Gigli GL. Epilepsy and episodic ataxia type 2: family study and review of the literature. J Neurol 2021; 268:4296-4302. [PMID: 33983550 DOI: 10.1007/s00415-021-10555-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022]
Abstract
Episodic ataxia type 2 (EA2) is a hereditary disorder characterized by paroxysmal attacks of ataxia, vertigo and nausea, due to mutations in the CACNA1A gene, which encodes for α1 subunit of the P/Q-type voltage-gated Ca2+ channel (CaV2.1). Other manifestations may be associated to CACNA1A mutations, such as migraine and epilepsy. The correlation between episodic ataxia and epilepsy is often underestimated and misdiagnosed. Clinical presentation of EA2 varies among patients and within the same family, and the same genetic mutation can lead to different clinical phenotypes. We herewith describe an Italian family presenting with typical EA2 and, in two of the family members (patients II.3 and III.1), epileptic seizures. The sequencing revealed a heterozygous deletion of 6 nucleotides in exon 28 of CACNA1A gene, present in all affected patients. Evidence suggests that mutations of CACNA1A, conferring a loss/reduction of CaV2.1 function, lead to an increase of thalamocortical excitation that contributes to epileptiform discharges. Our description highlights intra-family variability of EA2 phenotype and suggests that mutations in the CACNA1A gene should be suspected in individuals with focal or generalized epilepsy, associated with a family history of episodic ataxia.
Collapse
Affiliation(s)
- Lorenzo Verriello
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, 33100, Udine, Italy.
| | - Giada Pauletto
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, 33100, Udine, Italy
| | - Annacarmen Nilo
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Incoronata Lonigro
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Department of Laboratory Medicine, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Elena Betto
- Department of Laboratory Medicine, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Mariarosaria Valente
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Francesco Curcio
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Department of Laboratory Medicine, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Gian Luigi Gigli
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy.,Department of Mathematics, Informatics and Physics (DMIF), University of Udine, Udine, Italy
| |
Collapse
|
14
|
Global genetic deletion of Ca V3.3 channels facilitates anaesthetic induction and enhances isoflurane-sparing effects of T-type calcium channel blockers. Sci Rep 2020; 10:21510. [PMID: 33299036 PMCID: PMC7725806 DOI: 10.1038/s41598-020-78488-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/13/2020] [Indexed: 01/02/2023] Open
Abstract
We previously documented that the CaV3.3 isoform of T-type calcium channels (T-channels) is inhibited by clinically relevant concentrations of volatile anaesthetics, including isoflurane. However, little is understood about the functional role of CaV3.3 channels in anaesthetic-induced hypnosis and underlying neuronal oscillations. To address this issue, we used CaV3.3 knock-out (KO) mice and a panselective T-channel blocker 3,5-dichloro-N-[1-(2,2-dimethyltetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide (TTA-P2). We found that mutant mice injected with the vehicle showed faster induction of hypnosis than wild-type (WT) mice, while the percent isoflurane at which hypnosis and immobility occurred was not different between two genotypes. Furthermore, we found that TTA-P2 facilitated isoflurane induction of hypnosis in the CaV3.3 KO mice more robustly than in the WT mice. Isoflurane-induced hypnosis following injections of TTA-P2 was accompanied with more prominent delta and theta EEG oscillations in the mutant mice, and reached burst-suppression pattern earlier when compared to the WT mice. Our findings point to a relatively specific value of CaV3.3 channels in anaesthetic induced hypnosis. Furthermore, we propose that T-channel blockers may be further explored as a valuable adjunct to reducing the usage of potent volatile anaesthetics, thereby improving their safety.
Collapse
|
15
|
Timic Stamenic T, Feseha S, Valdez R, Zhao W, Klawitter J, Todorovic SM. Alterations in Oscillatory Behavior of Central Medial Thalamic Neurons Demonstrate a Key Role of CaV3.1 Isoform of T-Channels During Isoflurane-Induced Anesthesia. Cereb Cortex 2020; 29:4679-4696. [PMID: 30715245 DOI: 10.1093/cercor/bhz002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 11/14/2022] Open
Abstract
Although the central medial nucleus (CeM) of the thalamus is an essential part of the arousal system for sleep and anesthesia initiation, the precise mechanisms that regulate its activity are not well studied. We examined the role of CaV3.1 isoform of T-type calcium channels (T-channels) in the excitability and rhythmic activity of CeM neurons during isoflurane (ISO)-induced anesthesia by using mouse genetics and selective pharmacology. Patch-clamp recordings taken from acute brain slices revealed that CaV3.1 channels in CeM are inhibited by prototypical volatile anesthetic ISO (250 and 500 μM) and selective T-channels blocker 3,5-dichloro-N-[1-(2,2-dimethyl-tetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide (TTA-P2). Both TTA-P2 and ISO attenuated tonic and burst firing modes, and hyperpolarized CeM neurons from wild type (WT) mice. These effects were greatly diminished or abolished in CaV3.1 null mice. Our ensuing in vivo local field potential (LFP) recordings from CeM indicated that the ability of TTA-P2 and anesthetic concentrations of ISO to promote δ oscillation was substantially weakened in CaV3.1 null mice. Furthermore, escalating ISO concentrations induced stronger burst-suppression LFP pattern in mutant than in WT mice. Our results demonstrate for the first time the importance of CaV3.1 channels in thalamocortical oscillations from the non-specific thalamic nuclei that underlie clinically important effects of ISO.
Collapse
Affiliation(s)
- Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Feseha
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Robert Valdez
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Wanzhu Zhao
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jost Klawitter
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,Neuroscience Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
16
|
Novel Missense CACNA1G Mutations Associated with Infantile-Onset Developmental and Epileptic Encephalopathy. Int J Mol Sci 2020; 21:ijms21176333. [PMID: 32878331 PMCID: PMC7503748 DOI: 10.3390/ijms21176333] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/29/2020] [Accepted: 08/29/2020] [Indexed: 11/17/2022] Open
Abstract
The CACNA1G gene encodes the low-voltage-activated Cav3.1 channel, which is expressed in various areas of the CNS, including the cerebellum. We studied two missense CACNA1G variants, p.L208P and p.L909F, and evaluated the relationships between the severity of Cav3.1 dysfunction and the clinical phenotype. The presentation was of a developmental and epileptic encephalopathy without evident cerebellar atrophy. Both patients exhibited axial hypotonia, developmental delay, and severe to profound cognitive impairment. The patient with the L909F mutation had initially refractory seizures and cerebellar ataxia, whereas the L208P patient had seizures only transiently but was overall more severely affected. In transfected mammalian cells, we determined the biophysical characteristics of L208P and L909F variants, relative to the wild-type channel and a previously reported gain-of-function Cav3.1 variant. The L208P mutation shifted the activation and inactivation curves to the hyperpolarized direction, slowed the kinetics of inactivation and deactivation, and reduced the availability of Ca2+ current during repetitive stimuli. The L909F mutation impacted channel function less severely, resulting in a hyperpolarizing shift of the activation curve and slower deactivation. These data suggest that L909F results in gain-of-function, whereas L208P exhibits mixed gain-of-function and loss-of-function effects due to opposing changes in the biophysical properties. Our study expands the clinical spectrum associated with CACNA1G mutations, corroborating further the causal association with distinct complex phenotypes.
Collapse
|
17
|
Miao QL, Herlitze S, Mark MD, Noebels JL. Adult loss of Cacna1a in mice recapitulates childhood absence epilepsy by distinct thalamic bursting mechanisms. Brain 2020; 143:161-174. [PMID: 31800012 PMCID: PMC6935748 DOI: 10.1093/brain/awz365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/09/2019] [Accepted: 09/29/2019] [Indexed: 12/15/2022] Open
Abstract
Inborn errors of CACNA1A-encoded P/Q-type calcium channels impair synaptic transmission, producing early and lifelong neurological deficits, including childhood absence epilepsy, ataxia and dystonia. Whether these impairments owe their pathologies to defective channel function during the critical period for thalamic network stabilization in immature brain remains unclear. Here we show that mice with tamoxifen-induced adult-onset ablation of P/Q channel alpha subunit (iKOp/q) display identical patterns of dysfunction, replicating the inborn loss-of-function phenotypes and, therefore demonstrate that these neurological defects do not rely upon developmental abnormality. Unexpectedly, unlike the inborn model, the adult-onset pattern of excitability changes believed to be pathogenic within the thalamic network is non-canonical. Specifically, adult ablation of P/Q channels does not promote Cacna1g-mediated burst firing or T-type calcium current (IT) in the thalamocortical relay neurons; however, burst firing in thalamocortical relay neurons remains essential as iKOp/q mice generated on a Cacna1g deleted background show substantially diminished seizure generation. Moreover, in thalamic reticular nucleus neurons, burst firing is impaired accompanied by attenuated IT. Interestingly, inborn deletion of thalamic reticular nucleus-enriched, human childhood absence epilepsy-linked gene Cacna1h in iKOp/q mice reduces thalamic reticular nucleus burst firing and promotes rather than reduces seizure, indicating an epileptogenic role for loss-of-function Cacna1h gene variants reported in human childhood absence epilepsy cases. Together, our results demonstrate that P/Q channels remain critical for maintaining normal thalamocortical oscillations and motor control in the adult brain, and suggest that the developmental plasticity of membrane currents regulating pathological rhythmicity is both degenerate and age-dependent.
Collapse
Affiliation(s)
- Qing-Long Miao
- Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston TX, USA
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr University of Bochum, Bochum, Germany
| | - Melanie D Mark
- Department of Zoology and Neurobiology, Ruhr University of Bochum, Bochum, Germany
| | - Jeffrey L Noebels
- Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX, USA
| |
Collapse
|
18
|
Teplov IY, Tuleukhanov ST, Zinchenko VP. Regulation of Action Potential Frequency and Amplitude by T-type Ca2+ Channel During Spontaneous Synchronous Activity of Hippocampal Neurons. Biophysics (Nagoya-shi) 2018. [DOI: 10.1134/s0006350918040206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
19
|
Abstract
Epilepsy, characterized by spontaneous recurrent seizures (SRS), is a serious and common neurological disorder afflicting an estimated 1% of the population worldwide. Animal experiments, especially those utilizing small laboratory rodents, remain essential to understanding the fundamental mechanisms underlying epilepsy and to prevent, diagnose, and treat this disease. While much attention has been focused on epileptogenesis in animal models of epilepsy, there is little discussion on SRS, the hallmark of epilepsy. This is in part due to the technical difficulties of rigorous SRS detection. In this review, we comprehensively summarize both genetic and acquired models of SRS and discuss the methodology used to monitor and detect SRS in mice and rats.
Collapse
Affiliation(s)
- Bin Gu
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Katherine A Dalton
- Psychology & Neuroscience Program, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
20
|
Oyrer J, Maljevic S, Scheffer IE, Berkovic SF, Petrou S, Reid CA. Ion Channels in Genetic Epilepsy: From Genes and Mechanisms to Disease-Targeted Therapies. Pharmacol Rev 2018; 70:142-173. [PMID: 29263209 PMCID: PMC5738717 DOI: 10.1124/pr.117.014456] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/02/2017] [Indexed: 12/19/2022] Open
Abstract
Epilepsy is a common and serious neurologic disease with a strong genetic component. Genetic studies have identified an increasing collection of disease-causing genes. The impact of these genetic discoveries is wide reaching-from precise diagnosis and classification of syndromes to the discovery and validation of new drug targets and the development of disease-targeted therapeutic strategies. About 25% of genes identified in epilepsy encode ion channels. Much of our understanding of disease mechanisms comes from work focused on this class of protein. In this study, we review the genetic, molecular, and physiologic evidence supporting the pathogenic role of a number of different voltage- and ligand-activated ion channels in genetic epilepsy. We also review proposed disease mechanisms for each ion channel and highlight targeted therapeutic strategies.
Collapse
Affiliation(s)
- Julia Oyrer
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Ingrid E Scheffer
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Samuel F Berkovic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia (J.O., S.M., I.E.S., S.P., C.A.R.); Department of Medicine, Austin Health, University of Melbourne, Heidelberg West, Melbourne, Australia (I.E.S., S.F.B.); and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia (I.E.S.)
| |
Collapse
|
21
|
Schneider T, Alpdogan S, Hescheler J, Neumaier F. In vitro and in vivo phosphorylation of the Ca v2.3 voltage-gated R-type calcium channel. Channels (Austin) 2018; 12:326-334. [PMID: 30165790 PMCID: PMC6986797 DOI: 10.1080/19336950.2018.1516984] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
During the recording of whole cell currents from stably transfected HEK-293 cells, the decline of currents carried by the recombinant human Cav2.3+β3 channel subunits is related to adenosine triphosphate (ATP) depletion after rupture of the cells. It reduces the number of functional channels and leads to a progressive shift of voltage-dependent gating to more negative potentials (Neumaier F., et al., 2018). Both effects can be counteracted by hydrolysable ATP, whose protective action is almost completely prevented by inhibition of serine/threonine but not tyrosine or lipid kinases. These findings indicate that ATP promotes phosphorylation of either the channel or an associated protein, whereas dephosphorylation during cell dialysis results in run-down. Protein phosphorylation is required for Cav2.3 channel function and could directly influence the normal features of current carried by these channels. Therefore, results from in vitro and in vivo phosphorylation of Cav2.3 are summarized to come closer to a functional analysis of structural variations in Cav2.3 splice variants.
Collapse
Affiliation(s)
- T. Schneider
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, Cologne, Germany
| | - S. Alpdogan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, Cologne, Germany
| | - J. Hescheler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, Cologne, Germany
| | - F. Neumaier
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, Cologne, Germany
| |
Collapse
|
22
|
Bezençon O, Heidmann B, Siegrist R, Stamm S, Richard S, Pozzi D, Corminboeuf O, Roch C, Kessler M, Ertel EA, Reymond I, Pfeifer T, de Kanter R, Toeroek-Schafroth M, Moccia LG, Mawet J, Moon R, Rey M, Capeleto B, Fournier E. Discovery of a Potent, Selective T-type Calcium Channel Blocker as a Drug Candidate for the Treatment of Generalized Epilepsies. J Med Chem 2017; 60:9769-9789. [PMID: 29116786 DOI: 10.1021/acs.jmedchem.7b01236] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We report here the discovery and pharmacological characterization of N-(1-benzyl-1H-pyrazol-3-yl)-2-phenylacetamide derivatives as potent, selective, brain-penetrating T-type calcium channel blockers. Optimization focused mainly on solubility, brain penetration, and the search for an aminopyrazole metabolite that would be negative in an Ames test. This resulted in the preparation and complete characterization of compound 66b (ACT-709478), which has been selected as a clinical candidate.
Collapse
Affiliation(s)
- Olivier Bezençon
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Bibia Heidmann
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Romain Siegrist
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Simon Stamm
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Sylvia Richard
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Davide Pozzi
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Olivier Corminboeuf
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Catherine Roch
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Melanie Kessler
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Eric A Ertel
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Isabelle Reymond
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Thomas Pfeifer
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Ruben de Kanter
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Michael Toeroek-Schafroth
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Luca G Moccia
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Jacques Mawet
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Richard Moon
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Markus Rey
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Bruno Capeleto
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Elvire Fournier
- Chemistry, Biology and Pharmacology & Pre-clinical Development, Drug Discovery, Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| |
Collapse
|
23
|
McArthur JR, Motin L, Gleeson EC, Spiller S, Lewis RJ, Duggan PJ, Tuck KL, Adams DJ. Inhibition of human N- and T-type calcium channels by an ortho-phenoxyanilide derivative, MONIRO-1. Br J Pharmacol 2017; 175:2284-2295. [PMID: 28608537 DOI: 10.1111/bph.13910] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/24/2017] [Accepted: 06/05/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Voltage-gated calcium channels are involved in nociception in the CNS and in the periphery. N-type (Cav 2.2) and T-type (Cav 3.1, Cav 3.2 and Cav 3.3) voltage-gated calcium channels are particularly important in studying and treating pain and epilepsy. EXPERIMENTAL APPROACH In this study, whole-cell patch clamp electrophysiology was used to assess the potency and mechanism of action of a novel ortho-phenoxylanilide derivative, MONIRO-1, against a panel of voltage-gated calcium channels including Cav 1.2, Cav 1.3, Cav 2.1, Cav 2.2, Cav 2.3, Cav 3.1, Cav 3.2 and Cav 3.3. KEY RESULTS MONIRO-1 was 5- to 20-fold more potent at inhibiting human T-type calcium channels, hCav 3.1, hCav 3.2 and hCav 3.3 (IC50 : 3.3 ± 0.3, 1.7 ± 0.1 and 7.2 ± 0.3 μM, respectively) than N-type calcium channel, hCav 2.2 (IC50 : 34.0 ± 3.6 μM). It interacted with L-type calcium channels Cav 1.2 and Cav 1.3 with significantly lower potency (IC50 > 100 μM) and did not inhibit hCav 2.1 or hCav 2.3 channels at concentrations as high as 100 μM. State- and use-dependent inhibition of hCav 2.2 channels was observed, whereas stronger inhibition occurred at high stimulation frequencies for hCav 3.1 channels suggesting a different mode of action between these two channels. CONCLUSIONS AND IMPLICATIONS Selectivity, potency, reversibility and multi-modal effects distinguish MONIRO-1 from other low MW inhibitors acting on Cav channels involved in pain and/or epilepsy pathways. High-frequency firing increased the affinity for MONIRO-1 for both hCav 2.2 and hCav 3.1 channels. Such Cav channel modulators have potential clinical use in the treatment of epilepsies, neuropathic pain and other nociceptive pathophysiologies. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Health Innovations Research Institute, RMIT University, Melbourne, VIC, Australia
| | - Leonid Motin
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Health Innovations Research Institute, RMIT University, Melbourne, VIC, Australia
| | - Ellen C Gleeson
- CSIRO Manufacturing, Bag 10, Clayton South, VIC, Australia.,School of Chemistry, Monash University, Clayton, VIC, Australia
| | - Sandro Spiller
- School of Chemistry, Monash University, Clayton, VIC, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Peter J Duggan
- CSIRO Manufacturing, Bag 10, Clayton South, VIC, Australia.,School of Chemical and Physical Sciences, Flinders University, Adelaide, SA, Australia
| | - Kellie L Tuck
- School of Chemistry, Monash University, Clayton, VIC, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Health Innovations Research Institute, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Wormuth C, Lundt A, Henseler C, Müller R, Broich K, Papazoglou A, Weiergräber M. Review: Ca v2.3 R-type Voltage-Gated Ca 2+ Channels - Functional Implications in Convulsive and Non-convulsive Seizure Activity. Open Neurol J 2016; 10:99-126. [PMID: 27843503 PMCID: PMC5080872 DOI: 10.2174/1874205x01610010099] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/16/2016] [Accepted: 06/24/2016] [Indexed: 11/22/2022] Open
Abstract
Background: Researchers have gained substantial insight into mechanisms of synaptic transmission, hyperexcitability, excitotoxicity and neurodegeneration within the last decades. Voltage-gated Ca2+ channels are of central relevance in these processes. In particular, they are key elements in the etiopathogenesis of numerous seizure types and epilepsies. Earlier studies predominantly targeted on Cav2.1 P/Q-type and Cav3.2 T-type Ca2+ channels relevant for absence epileptogenesis. Recent findings bring other channels entities more into focus such as the Cav2.3 R-type Ca2+ channel which exhibits an intriguing role in ictogenesis and seizure propagation. Cav2.3 R-type voltage gated Ca2+ channels (VGCC) emerged to be important factors in the pathogenesis of absence epilepsy, human juvenile myoclonic epilepsy (JME), and cellular epileptiform activity, e.g. in CA1 neurons. They also serve as potential target for various antiepileptic drugs, such as lamotrigine and topiramate. Objective: This review provides a summary of structure, function and pharmacology of VGCCs and their fundamental role in cellular Ca2+ homeostasis. We elaborate the unique modulatory properties of Cav2.3 R-type Ca2+ channels and point to recent findings in the proictogenic and proneuroapoptotic role of Cav2.3 R-type VGCCs in generalized convulsive tonic–clonic and complex-partial hippocampal seizures and its role in non-convulsive absence like seizure activity. Conclusion: Development of novel Cav2.3 specific modulators can be effective in the pharmacological treatment of epilepsies and other neurological disorders.
Collapse
Affiliation(s)
- Carola Wormuth
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Andreas Lundt
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Christina Henseler
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Ralf Müller
- Department of Psychiatry and Psychotherapy, University of Cologne, Faculty of Medicine, Cologne, Germany
| | - Karl Broich
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Anna Papazoglou
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Marco Weiergräber
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| |
Collapse
|
25
|
Dissecting the Role of P/Q-Type Calcium Channels in Corticothalamic Circuit Dysfunction and Absence Epilepsy. J Neurosci 2016; 36:5677-9. [PMID: 27225758 DOI: 10.1523/jneurosci.0753-16.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022] Open
|
26
|
Isolated P/Q Calcium Channel Deletion in Layer VI Corticothalamic Neurons Generates Absence Epilepsy. J Neurosci 2016; 36:405-18. [PMID: 26758833 DOI: 10.1523/jneurosci.2555-15.2016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Generalized spike-wave seizures involving abnormal synchronization of cortical and underlying thalamic circuitry represent a major category of childhood epilepsy. Inborn errors of Cacna1a, the P/Q-type voltage-gated calcium channel α subunit gene, expressed throughout the brain destabilize corticothalamic rhythmicity and produce this phenotype. To determine the minimal cellular lesion required for this network disturbance, we used neurotensin receptor 1 (Ntsr1) cre-driver mice to ablate floxed Cacna1a in layer VI pyramidal neurons, which supply the sole descending cortical synaptic input to thalamocortical relay cells and reticular interneurons and activate intrathalamic circuits. Targeted Cacna1a ablation in layer VI cells resulted in mice that display a robust spontaneous spike-wave absence seizure phenotype accompanied by behavioral arrest and inhibited by ethosuximide. To verify the selectivity of the molecular lesion, we determined that P/Q subunit proteins were reduced in corticothalamic relay neuron terminal zones, and confirmed that P/Q-mediated glutamate release was reduced at these synapses. Spike-triggered exocytosis was preserved by N-type calcium channel rescue, demonstrating that evoked release at layer VI terminals relies on both P/Q and N-type channels. Whereas intrinsic excitability of the P/Q channel depleted layer VI neurons was unaltered, T-type calcium currents in the postsynaptic thalamic relay and reticular cells were dramatically elevated, favoring rebound bursting and seizure generation. We find that an early P/Q-type release defect, limited to synapses of a single cell-type within the thalamocortical circuit, is sufficient to remodel synchronized firing behavior and produce a stable generalized epilepsy phenotype. SIGNIFICANCE STATEMENT This study dissects a critical component of the corticothalamic circuit in spike-wave epilepsy and identifies the developmental importance of P/Q-type calcium channel-mediated presynaptic glutamate release at layer VI pyramidal neuron terminals. Genetic ablation of Cacna1a in layer VI neurons produced synchronous spike-wave discharges in the cortex and thalamus that were inhibited by ethosuximide. These mice also displayed N-type calcium channel compensation at descending thalamic synapses, and consistent with other spike-wave models increased low-threshold T-type calcium currents within postsynaptic thalamic relay and reticular neurons. These results demonstrate, for the first time, that preventing the developmental homeostatic switch from loose to tightly coupled synaptic release at a single class of deep layer cortical excitatory output neurons results in generalized spike-wave epilepsy.
Collapse
|
27
|
Calhoun JD, Hawkins NA, Zachwieja NJ, Kearney JA. Cacna1g is a genetic modifier of epilepsy caused by mutation of voltage-gated sodium channel Scn2a. Epilepsia 2016; 57:e103-7. [PMID: 27112236 DOI: 10.1111/epi.13390] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2016] [Indexed: 01/02/2023]
Abstract
More than 1,200 mutations in neuronal voltage-gated sodium channel (VGSC) genes have been identified in patients with several epilepsy syndromes. A common feature of genetic epilepsies is variable expressivity among individuals with the same mutation. The Scn2a(Q54) transgenic mouse model has a mutation in Scn2a that results in spontaneous epilepsy. Scn2a(Q54) phenotype severity varies depending on the genetic strain background, making it a useful model for identifying and characterizing epilepsy modifier genes. Scn2a(Q54) mice on the [C57BL/6JxSJL/J]F1 background exhibit earlier seizure onset, elevated spontaneous seizure frequency, and decreased survival compared to Scn2a(Q54) mice congenic on the C57BL/6J strain. Genetic mapping and RNA-Seq analysis identified Cacna1g as a candidate modifier gene at the Moe1 locus, which influences Scn2a(Q54) phenotype severity. In this study, we evaluated the modifier potential of Cacna1g, encoding the Cav3.1 voltage-gated calcium channel, by testing whether transgenic alteration of Cacna1g expression modifies severity of the Scn2a(Q54) seizure phenotype. Scn2a(Q54) mice exhibited increased spontaneous seizure frequency with elevated Cacna1g expression and decreased seizure frequency with decreased Cacna1g expression. These results provide support for Cacna1g as an epilepsy modifier gene and suggest that modulation of Cav3.1 may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jeffrey D Calhoun
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, U.S.A.,Department of Medicine, Vanderbilt University, Nashville, Tennessee, U.S.A
| | - Nicole A Hawkins
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, U.S.A.,Neuroscience Program, Vanderbilt University, Nashville, Tennessee, U.S.A
| | - Nicole J Zachwieja
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, U.S.A
| | - Jennifer A Kearney
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, U.S.A.,Department of Medicine, Vanderbilt University, Nashville, Tennessee, U.S.A
| |
Collapse
|
28
|
Abstract
A central theme in the quest to unravel the genetic basis of epilepsy has been the effort to elucidate the roles played by inherited defects in ion channels. The ubiquitous expression of voltage-gated calcium channels (VGCCs) throughout the central nervous system (CNS), along with their involvement in fundamental processes, such as neuronal excitability and synaptic transmission, has made them attractive candidates. Recent insights provided by the identification of mutations in the P/Q-type calcium channel in humans and rodents with epilepsy and the finding of thalamic T-type calcium channel dysfunction in the absence of seizures have raised expectations of a causal role of calcium channels in the polygenic inheritance of idiopathic epilepsy. In this review, we consider how genetic variation in neuronal VGCCs may influence the development of epilepsy.
Collapse
Affiliation(s)
- Sanjeev Rajakulendran
- UCL-Institute of Neurology, MRC Centre for Neuromuscular Diseases, Queen Square, London WC1N 3BG, United Kingdom
| | - Michael G Hanna
- UCL-Institute of Neurology, MRC Centre for Neuromuscular Diseases, Queen Square, London WC1N 3BG, United Kingdom
| |
Collapse
|
29
|
Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov 2015; 15:19-34. [DOI: 10.1038/nrd.2015.5] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
30
|
Kim CH. Cav3.1 T-type calcium channel modulates the epileptogenicity of hippocampal seizures in the kainic acid-induced temporal lobe epilepsy model. Brain Res 2015; 1622:204-16. [PMID: 26111648 DOI: 10.1016/j.brainres.2015.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 10/23/2022]
Abstract
The molecular mechanism of temporal lobe epilepsy has not been clearly identified. T-type calcium channels play a role in burst firing in neurons and have been implicated in several seizure models. In this study, the role of Cav3.1 T-type (α1G) calcium channel has been investigated in the kainic acid (KA)-induced temporal lobe epilepsy model (TLE) by using conventional α1G knock-out (ko) mice. After intraperitoneal (i.p.) administration or intrahippocampal injection of KA, depth hippocampal and cortical electroencephalogram (EEG) and behavioral monitoring were recorded, and timm and Nissl staining of brain sections were made later. Seizure was mainly identified by EEG signals, rather than behaviorally, with analytic criteria. During the acute status epilepticus (SE) period, both the duration and the frequency of hippocampal seizures were significantly reduced and increased, respectively, in αlG ko mice compared to those of wild type mice. Epileptogenicity, the total period of seizures (hr(-1)), was also significantly reduced in α1G ko mice. However, the latency of seizure occurrence was not significantly different between wild type and ko mice. These differential effects were not observed in cortical seizures. Furthermore, the injection of KA caused a strong increase in δ rhythm power spectrum density (PSD) of EEG in αlG ko mice compared to that in wild type mice. The results with conventional ko mice indicate that α1G T-type calcium channel plays a modulatory role in the duration and frequency of hippocampal seizures as well as the epileptogenicity of KA-induced TLE in mice, mostly during acute periods.
Collapse
Affiliation(s)
- Chong-Hyun Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science & Technology, Seoul 136-791, Republic of Korea; Department of Neuroscience, Korea University of Science & Technology, Daejeon 305-333, Republic of Korea.
| |
Collapse
|
31
|
Altered thalamocortical rhythmicity and connectivity in mice lacking CaV3.1 T-type Ca2+ channels in unconsciousness. Proc Natl Acad Sci U S A 2015; 112:7839-44. [PMID: 26056284 DOI: 10.1073/pnas.1420983112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In unconscious status (e.g., deep sleep and anesthetic unconsciousness) where cognitive functions are not generated there is still a significant level of brain activity present. Indeed, the electrophysiology of the unconscious brain is characterized by well-defined thalamocortical rhythmicity. Here we address the ionic basis for such thalamocortical rhythms during unconsciousness. In particular, we address the role of CaV3.1 T-type Ca(2+) channels, which are richly expressed in thalamic neurons. Toward this aim, we examined the electrophysiological and behavioral phenotypes of mice lacking CaV3.1 channels (CaV3.1 knockout) during unconsciousness induced by ketamine or ethanol administration. Our findings indicate that CaV3.1 KO mice displayed attenuated low-frequency oscillations in thalamocortical loops, especially in the 1- to 4-Hz delta band, compared with control mice (CaV3.1 WT). Intriguingly, we also found that CaV3.1 KO mice exhibited augmented high-frequency oscillations during unconsciousness. In a behavioral measure of unconsciousness dynamics, CaV3.1 KO mice took longer to fall into the unconscious state than controls. In addition, such unconscious events had a shorter duration than those of control mice. The thalamocortical interaction level between mediodorsal thalamus and frontal cortex in CaV3.1 KO mice was significantly lower, especially for delta band oscillations, compared with that of CaV3.1 WT mice, during unconsciousness. These results suggest that the CaV3.1 channel is required for the generation of a given set of thalamocortical rhythms during unconsciousness. Further, that thalamocortical resonant neuronal activity supported by this channel is important for the control of vigilance states.
Collapse
|
32
|
Lee HJ, Son Y, Kim J, Lee CJ, Yoon ES, Cho IJ. A multichannel neural probe with embedded microfluidic channels for simultaneous in vivo neural recording and drug delivery. LAB ON A CHIP 2015; 15:1590-7. [PMID: 25651943 DOI: 10.1039/c4lc01321b] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Multi-functional neural probes integrated with various stimulation modalities are becoming essential tools in neuroscience to study the brain more effectively. In this paper, we present a new multi-functional neural probe that allows chemical stimulation through drug delivery and simultaneous recording of individual neuron signals through a microelectrode array. By embedding microchannels in silicon using a proposed glass reflow process, we successfully fabricated 40 μm thick silicon neural probes suitable for small animal experiments. The electrochemical impedance spectroscopy confirms that impedance of iridium microelectrodes is low enough (<1 MΩ at 1 kHz) to measure neural signals. Flow rate characterization in a 0.9% w/v agarose gel shows the capability to deliver a small volume of drugs (<1 μl) at a controlled flow rate. We demonstrate the viability and potential of this new probe by conducting in vivo experiments on mice. Because of the proposed compact structure, both action potentials of individual neurons and local field potentials (LFP) at the thalamus region of a mouse brain were successfully detected with a noise level of ~30 μVpp. Furthermore, we successfully induced absence seizure by injecting seizure-inducing drugs (baclofen) at a local target region and observed distinctive changes in neural signal patterns. Specifically, spike-wave discharge (SWD), which is an indicative signal pattern of absence seizure, was successfully recorded. These signals were also directly compared to SWD detected after inducing absence seizure through direct injection of baclofen through the abdomen. This work demonstrates the potential of our multi-functional neural probes for use in effective investigation of brain functions and disorders by using widely available mouse models.
Collapse
Affiliation(s)
- Hyunjoo J Lee
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
33
|
Powell KL, Cain SM, Snutch TP, O'Brien TJ. Low threshold T-type calcium channels as targets for novel epilepsy treatments. Br J Clin Pharmacol 2015; 77:729-39. [PMID: 23834404 DOI: 10.1111/bcp.12205] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 07/02/2013] [Indexed: 12/21/2022] Open
Abstract
Low voltage-activated T-type calcium channels were originally cloned in the 1990s and much research has since focused on identifying the physiological roles of these channels in health and disease states. T-type calcium channels are expressed widely throughout the brain and peripheral tissues, and thus have been proposed as therapeutic targets for a variety of diseases such as epilepsy, insomnia, pain, cancer and hypertension. This review discusses the literature concerning the role of T-type calcium channels in physiological and pathological processes related to epilepsy. T-type calcium channels have been implicated in pathology of both the genetic and acquired epilepsies and several anti-epileptic drugs (AEDs) in clinical use are known to suppress seizures via inhibition of T-type calcium channels. Despite the fact that more than 15 new AEDs have become clinically available over the past 20 years at least 30% of epilepsy patients still fail to achieve seizure control, and many patients experience unwanted side effects. Furthermore there are no treatments that prevent the development of epilepsy or mitigate the epileptic state once established. Therefore there is an urgent need for the development of new AEDs that are effective in patients with drug resistant epilepsy, are anti-epileptogenic and are better tolerated. We also review the mechanisms of action of the current AEDs with known effects on T-type calcium channels and discuss novel compounds that are being investigated as new treatments for epilepsy.
Collapse
Affiliation(s)
- Kim L Powell
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
34
|
Rebound burst firing in the reticular thalamus is not essential for pharmacological absence seizures in mice. Proc Natl Acad Sci U S A 2014; 111:11828-33. [PMID: 25071191 DOI: 10.1073/pnas.1408609111] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Intrinsic burst and rhythmic burst discharges (RBDs) are elicited by activation of T-type Ca(2+) channels in the thalamic reticular nucleus (TRN). TRN bursts are believed to be critical for generation and maintenance of thalamocortical oscillations, leading to the spike-and-wave discharges (SWDs), which are the hallmarks of absence seizures. We observed that the RBDs were completely abolished, whereas tonic firing was significantly increased, in TRN neurons from mice in which the gene for the T-type Ca(2+) channel, CaV3.3, was deleted (CaV3.3(-/-)). Contrary to expectations, there was an increased susceptibility to drug-induced SWDs both in CaV3.3(-/-) mice and in mice in which the CaV3.3 gene was silenced predominantly in the TRN. CaV3.3(-/-) mice also showed enhanced inhibitory synaptic drive onto TC neurons. Finally, a double knockout of both CaV3.3 and CaV3.2, which showed complete elimination of burst firing and RBDs in TRN neurons, also displayed enhanced drug-induced SWDs and absence seizures. On the other hand, tonic firing in the TRN was increased in these mice, suggesting that increased tonic firing in the TRN may be sufficient for drug-induced SWD generation in the absence of burst firing. These results call into question the role of burst firing in TRN neurons in the genesis of SWDs, calling for a rethinking of the mechanism for absence seizure induction.
Collapse
|
35
|
Thalamocortical neurons display suppressed burst-firing due to an enhanced Ih current in a genetic model of absence epilepsy. Pflugers Arch 2014; 467:1367-82. [PMID: 24953239 PMCID: PMC4435665 DOI: 10.1007/s00424-014-1549-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 05/28/2014] [Accepted: 05/30/2014] [Indexed: 10/25/2022]
Abstract
Burst-firing in distinct subsets of thalamic relay (TR) neurons is thought to be a key requirement for the propagation of absence seizures. However, in the well-regarded Genetic Absence Epilepsy Rats from Strasbourg (GAERS) model as yet there has been no link described between burst-firing in TR neurons and spike-and-wave discharges (SWDs). GAERS ventrobasal (VB) neurons are a specific subset of TR neurons that do not normally display burst-firing during absence seizures in the GAERS model, and here, we assessed the underlying relationship of VB burst-firing with Ih and T-type calcium currents between GAERS and non-epileptic control (NEC) animals. In response to 200-ms hyperpolarizing current injections, adult epileptic but not pre-epileptic GAERS VB neurons displayed suppressed burst-firing compared to NEC. In response to longer duration 1,000-ms hyperpolarizing current injections, both pre-epileptic and epileptic GAERS VB neurons required significantly more hyperpolarizing current injection to burst-fire than those of NEC animals. The current density of the Hyperpolarization and Cyclic Nucleotide-activated (HCN) current (Ih) was found to be increased in GAERS VB neurons, and the blockade of Ih relieved the suppressed burst-firing in both pre-epileptic P15-P20 and adult animals. In support, levels of HCN-1 and HCN-3 isoform channel proteins were increased in GAERS VB thalamic tissue. T-type calcium channel whole-cell currents were found to be decreased in P7-P9 GAERS VB neurons, and also noted was a decrease in CaV3.1 mRNA and protein levels in adults. Z944, a potent T-type calcium channel blocker with anti-epileptic properties, completely abolished hyperpolarization-induced VB burst-firing in both NEC and GAERS VB neurons.
Collapse
|
36
|
Zhu Z, Zeng XH, Turecek J, Han VZ, Welsh JP. RNA interference of GluN1 inhibits neuronal rhythmogenesis in the adult inferior olive. J Mol Neurosci 2014; 55:416-29. [PMID: 24930901 DOI: 10.1007/s12031-014-0353-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/10/2014] [Indexed: 11/25/2022]
Abstract
RNA interference (RNAi) to knockdown N-methyl-D-aspartate receptor (NMDAR) function is being investigated to address disorders associated with pathological brain rhythms. A motivating finding has been that pharmacological block of NMDARs inhibited oscillations in neuronal membrane potential that entrain rhythmic bursts of action potentials. To determine whether transient effects of NMDAR antagonist drugs to inhibit neuronal rhythmicity can be stably induced with genetic specificity, we examined the effects of RNAi of GluN1 protein on the subthreshold oscillations (STOs) of neurons in the inferior olive (IO), a pacemaking nucleus necessary for motor and cognitive timing. Western blot of dissociated neurons demonstrated 90% knockdown of GluN1 after a strong in vivo transduction by a dual-microRNA lentiviral vector. GluN1 RNAi in whole-cell-patched IO neurons blocked both membrane depolarization and STOs typically induced by NMDAR activation for up to 54 days without affecting input resistance, membrane capacitance, action potential firing, high-threshold Ca(2+) spikes, the hyperpolarization-activated current Ih, or the activation of the low-threshold Ca(2+) current I(T). Although an off-target effect on Cav3 expression was ruled out also by BlastN query, we found that GluN1 RNAi chronically eliminated I(T)-dependent STOs at resting membrane potential, well below the activation threshold of the NMDAR channel. In the context of a recent report showing that NMDAR activation induces STOs as it strengthens electrical coupling, the long-term block of STOs by GluN1 RNAi may relate to the loss of an essential support mechanism. Lentivector-mediated RNAi of GluN1 provides a novel technique for future investigations of NMDAR involvement in electrical oscillations and behavior.
Collapse
Affiliation(s)
- Zhiyi Zhu
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA, 98101, USA
| | | | | | | | | |
Collapse
|
37
|
Regionally specific expression of high-voltage-activated calcium channels in thalamic nuclei of epileptic and non-epileptic rats. Mol Cell Neurosci 2014; 61:110-22. [PMID: 24914823 DOI: 10.1016/j.mcn.2014.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 05/30/2014] [Accepted: 06/05/2014] [Indexed: 11/21/2022] Open
Abstract
The polygenic origin of generalized absence epilepsy results in dysfunction of ion channels that allows the switch from physiological asynchronous to pathophysiological highly synchronous network activity. Evidence from rat and mouse models of absence epilepsy indicates that altered Ca(2+) channel activity contributes to cellular and network alterations that lead to seizure activity. Under physiological circumstances, high voltage-activated (HVA) Ca(2+) channels are important in determining the thalamic firing profile. Here, we investigated a possible contribution of HVA channels to the epileptic phenotype using a rodent genetic model of absence epilepsy. In this study, HVA Ca(2+) currents were recorded from neurons of three different thalamic nuclei that are involved in both sensory signal transmission and rhythmic-synchronized activity during epileptic spike-and-wave discharges (SWD), namely the dorsal part of the lateral geniculate nucleus (dLGN), the ventrobasal thalamic complex (VB) and the reticular thalamic nucleus (NRT) of epileptic Wistar Albino Glaxo rats from Rijswijk (WAG/Rij) and non-epileptic August Copenhagen Irish (ACI) rats. HVA Ca(2+) current densities in dLGN neurons were significantly increased in epileptic rats compared with non-epileptic controls while other thalamic regions revealed no differences between the strains. Application of specific channel blockers revealed that the increased current was carried by L-type Ca(2+) channels. Electrophysiological evidence of increased L-type current correlated with up-regulated mRNA and protein expression of a particular L-type channel, namely Cav1.3, in dLGN of epileptic rats. No significant changes were found for other HVA Ca(2+) channels. Moreover, pharmacological inactivation of L-type Ca(2+) channels results in altered firing profiles of thalamocortical relay (TC) neurons from non-epileptic rather than from epileptic rats. While HVA Ca(2+) channels influence tonic and burst firing in ACI and WAG/Rij differently, it is discussed that increased Cav1.3 expression may indirectly contribute to increased robustness of burst firing and thereby the epileptic phenotype of absence epilepsy.
Collapse
|
38
|
Simms BA, Zamponi GW. Neuronal voltage-gated calcium channels: structure, function, and dysfunction. Neuron 2014; 82:24-45. [PMID: 24698266 DOI: 10.1016/j.neuron.2014.03.016] [Citation(s) in RCA: 456] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Voltage-gated calcium channels are the primary mediators of depolarization-induced calcium entry into neurons. There is great diversity of calcium channel subtypes due to multiple genes that encode calcium channel α1 subunits, coassembly with a variety of ancillary calcium channel subunits, and alternative splicing. This allows these channels to fulfill highly specialized roles in specific neuronal subtypes and at particular subcellular loci. While calcium channels are of critical importance to brain function, their inappropriate expression or dysfunction gives rise to a variety of neurological disorders, including, pain, epilepsy, migraine, and ataxia. This Review discusses salient aspects of voltage-gated calcium channel function, physiology, and pathophysiology.
Collapse
Affiliation(s)
- Brett A Simms
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
39
|
Chen Y, Parker WD, Wang K. The role of T-type calcium channel genes in absence seizures. Front Neurol 2014; 5:45. [PMID: 24847307 PMCID: PMC4023043 DOI: 10.3389/fneur.2014.00045] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/24/2014] [Indexed: 12/05/2022] Open
Abstract
The thalamic relay neurons, reticular thalamic nucleus, and neocortical pyramidal cells form a circuit that sustains oscillatory burst firing, and is regarded as the underlying mechanism of absence seizures. T-type calcium channels play a key role in this circuit. Here, we review the role of T-type calcium channel genes in the development of absence seizures, and emphasize gain or loss of function mutations, and other variations that alter both quantity and quality of transcripts, and methylation status of isoforms of T-type calcium channel proteins might be of equal importance in understanding the pathological mechanism of absence seizures.
Collapse
Affiliation(s)
- Yucai Chen
- University of Illinois at Chicago , Peoria, IL , USA
| | | | - Keling Wang
- Hebei Children Hospital , Shijiazhuang , China
| |
Collapse
|
40
|
Siwek ME, Müller R, Henseler C, Broich K, Papazoglou A, Weiergräber M. The CaV2.3 R-type voltage-gated Ca2+ channel in mouse sleep architecture. Sleep 2014; 37:881-92. [PMID: 24790266 DOI: 10.5665/sleep.3652] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Voltage-gated Ca(2+) channels (VGCCs) are key elements in mediating thalamocortical rhythmicity. Low-voltage activated (LVA) CaV 3 T-type Ca(2+) channels have been related to thalamic rebound burst firing and to generation of non-rapid eye movement (NREM) sleep. High-voltage activated (HVA) CaV 1 L-type Ca(2+) channels, on the opposite, favor the tonic mode of action associated with higher levels of vigilance. However, the role of the HVA Non-L-type CaV2.3 Ca(2+) channels, which are predominantly expressed in the reticular thalamic nucleus (RTN), still remains unclear. Recently, CaV2.3(-/-) mice were reported to exhibit altered spike-wave discharge (SWD)/absence seizure susceptibility supported by the observation that CaV2.3 mediated Ca(2+) influx into RTN neurons can trigger small-conductance Ca(2+)-activated K(+)-channel type 2 (SK2) currents capable of maintaining thalamic burst activity. Based on these studies we investigated the role of CaV2.3 R-type Ca(2+) channels in rodent sleep. METHODS The role of CaV2.3 Ca(2+) channels was analyzed in CaV2.3(-/-) mice and controls in both spontaneous and artificial urethane-induced sleep, using implantable video-EEG radiotelemetry. Data were analyzed for alterations in sleep architecture using sleep staging software and time-frequency analysis. RESULTS CaV2.3 deficient mice exhibited reduced wake duration and increased slow-wave sleep (SWS). Whereas mean sleep stage durations remained unchanged, the total number of SWS epochs was increased in CaV2.3(-/-) mice. Additional changes were observed for sleep stage transitions and EEG amplitudes. Furthermore, urethane-induced SWS mimicked spontaneous sleep results obtained from CaV2.3 deficient mice. Quantitative Real-time PCR did not reveal changes in thalamic CaV3 T-type Ca(2+) channel expression. The detailed mechanisms of SWS increase in CaV2.3(-/-) mice remain to be determined. CONCLUSIONS Low-voltage activated CaV2.3 R-type Ca(2+) channels in the thalamocortical loop and extra-thalamocortical circuitries substantially regulate rodent sleep architecture thus representing a novel potential target for pharmacological treatment of sleep disorders in the future.
Collapse
Affiliation(s)
| | - Ralf Müller
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | | | - Karl Broich
- Federal Institute for Drugs and Medical Devices, Bonn, BfArM, Germany
| | - Anna Papazoglou
- Federal Institute for Drugs and Medical Devices, Bonn, BfArM, Germany
| | - Marco Weiergräber
- Federal Institute for Drugs and Medical Devices, Bonn, BfArM, Germany ; Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| |
Collapse
|
41
|
Kopecky BJ, Liang R, Bao J. T-type calcium channel blockers as neuroprotective agents. Pflugers Arch 2014; 466:757-65. [PMID: 24563219 DOI: 10.1007/s00424-014-1454-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/15/2014] [Accepted: 01/19/2014] [Indexed: 01/12/2023]
Abstract
T-type calcium channels are expressed in many diverse tissues, including neuronal, cardiovascular, and endocrine. T-type calcium channels are known to play roles in the development, maintenance, and repair of these tissues but have also been implicated in disease when not properly regulated. Calcium channel blockers have been developed to treat various diseases and their use clinically is widespread due to both their efficacy as well as their safety. Aside from their established clinical applications, recent studies have suggested neuroprotective effects of T-type calcium channel blockers. Many of the current T-type calcium channel blockers could act on other molecular targets besides T-type calcium channels making it uncertain whether their neuroprotective effects are solely due to blocking of T-type calcium channels. In this review, we discuss these drugs as well as newly developed chemical compounds that are designed to be more selective for T-type calcium channels. We review in vitro and in vivo evidence of neuroprotective effects by these T-type calcium channel blockers. We conclude by discussing possible molecular mechanisms underlying the neuroprotective effects by T-type calcium channel blockers.
Collapse
Affiliation(s)
- Benjamin J Kopecky
- Department of Otolaryngology, Center for Aging, Washington University School of Medicine, 4560 Clayton Avenue, St. Louis, MO, 63110, USA
| | | | | |
Collapse
|
42
|
Paolicchi JM. The timing of pediatric epilepsy syndromes: what are the developmental triggers? Ann N Y Acad Sci 2014; 1304:45-51. [PMID: 24279892 DOI: 10.1111/nyas.12307] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pediatric epilepsy is characterized by multiple epilepsy syndromes with specific developmental triggers. They initiate spontaneously at critical periods of development and can just as spontaneously remit. Accompanying neurocognitive disabilities are often specific to the epileptic syndrome. Infantile or epileptic spasms have a very specific developmental window in the first year of life. Preceding the epilepsy, developmental arrest is common. The neurologic pathways underlying the development of spasms have been identified through PET scans as developmental abnormalities of serotonergic and GABAergic neurotransmitter systems in the brain stem and basal ganglia. Childhood absence epilepsy (CAE) and benign centrotemporal epilepsy syndrome (BECTS) are both known genetic epilepsy syndromes; they have a discrete onset in childhood with remission by puberty. In CAE, disturbances of specific calcium channels at key developmental stages lead to aberrant disruption of thalamocortical synchrony. Similarly, a complex interplay between brain development, maturation, and susceptibility genes underlies the seizures and the neurocognitive deficits of BECTS.
Collapse
|
43
|
T-type Ca2+ channels in absence epilepsy. Pflugers Arch 2014; 466:719-34. [DOI: 10.1007/s00424-014-1461-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/22/2014] [Indexed: 11/25/2022]
|
44
|
Maheshwari A, Noebels JL. Monogenic models of absence epilepsy: windows into the complex balance between inhibition and excitation in thalamocortical microcircuits. PROGRESS IN BRAIN RESEARCH 2014; 213:223-52. [PMID: 25194492 DOI: 10.1016/b978-0-444-63326-2.00012-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Absence epilepsy is a common disorder that arises in childhood and can be refractory to medical treatment. Single genetic mutations in mice, at times found in patients with absence epilepsy, provide the unique opportunity to bridge the gap between dysfunction at the genetic level and pathological oscillations within the thalamocortical circuit. Interestingly, unlike other forms of epilepsy, only genes related to ion channels have so far been linked to absence phenotypes. Here, we delineate a paradigm which attempts to unify the various monogenic models based on decades of research. While reviewing the particular impact of these individual mutations, we posit a framework involving fast feedforward disinhibition as one common mechanism that can lead to increased tonic inhibition in the cortex and/or thalamus. Enhanced tonic inhibition hyperpolarizes principal cells, deinactivates T-type calcium channels, and leads to reciprocal burst firing within the thalamocortical loop. We also review data from pharmacologic and polygenic models in light of this paradigm. Ultimately, many questions remain unanswered regarding the pathogenesis of absence epilepsy.
Collapse
Affiliation(s)
- Atul Maheshwari
- Department of Neurology, Developmental Neurogenetics Laboratory, Baylor College of Medicine Houston, TX, USA.
| | - Jeffrey L Noebels
- Department of Neurology, Developmental Neurogenetics Laboratory, Baylor College of Medicine Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
45
|
Sleep spindles are generated in the absence of T-type calcium channel-mediated low-threshold burst firing of thalamocortical neurons. Proc Natl Acad Sci U S A 2013; 110:20266-71. [PMID: 24282303 DOI: 10.1073/pnas.1320572110] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T-type Ca(2+) channels in thalamocortical (TC) neurons have long been considered to play a critical role in the genesis of sleep spindles, one of several TC oscillations. A classical model for TC oscillations states that reciprocal interaction between synaptically connected GABAergic thalamic reticular nucleus (TRN) neurons and glutamatergic TC neurons generates oscillations through T-type channel-mediated low-threshold burst firings of neurons in the two nuclei. These oscillations are then transmitted from TC neurons to cortical neurons, contributing to the network of TC oscillations. Unexpectedly, however, we found that both WT and KO mice for CaV3.1, the gene for T-type Ca(2+) channels in TC neurons, exhibit typical waxing-and-waning sleep spindle waves at a similar occurrence and with similar amplitudes and episode durations during non-rapid eye movement sleep. Single-unit recording in parallel with electroencephalography in vivo confirmed a complete lack of burst firing in the mutant TC neurons. Of particular interest, the tonic spike frequency in TC neurons was significantly increased during spindle periods compared with nonspindle periods in both genotypes. In contrast, no significant change in burst firing frequency between spindle and nonspindle periods was noted in the WT mice. Furthermore, spindle-like oscillations were readily generated within intrathalamic circuits composed solely of TRN and TC neurons in vitro in both the KO mutant and WT mice. Our findings call into question the essential role of low-threshold burst firings in TC neurons and suggest that tonic firing is important for the generation and propagation of spindle oscillations in the TC circuit.
Collapse
|
46
|
Abstract
Low-voltage-activated T-type Ca(2+) channels are widely expressed in various types of neurons. Once deinactivated by hyperpolarization, T-type channels are ready to be activated by a small depolarization near the resting membrane potential and, therefore, are optimal for regulating the excitability and electroresponsiveness of neurons under physiological conditions near resting states. Ca(2+) influx through T-type channels engenders low-threshold Ca(2+) spikes, which in turn trigger a burst of action potentials. Low-threshold burst firing has been implicated in the synchronization of the thalamocortical circuit during sleep and in absence seizures. It also has been suggested that T-type channels play an important role in pain signal transmission, based on their abundant expression in pain-processing pathways in peripheral and central neurons. In this review, we will describe studies on the role of T-type Ca(2+) channels in the physiological as well as pathological generation of brain rhythms in sleep, absence epilepsy, and pain signal transmission. Recent advances in studies of T-type channels in the control of cognition will also be briefly discussed.
Collapse
Affiliation(s)
- Eunji Cheong
- Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | | |
Collapse
|
47
|
Rossignol E, Kruglikov I, van den Maagdenberg AMJM, Rudy B, Fishell G. CaV 2.1 ablation in cortical interneurons selectively impairs fast-spiking basket cells and causes generalized seizures. Ann Neurol 2013; 74:209-22. [PMID: 23595603 DOI: 10.1002/ana.23913] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 04/03/2013] [Accepted: 04/12/2013] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Both the neuronal populations and mechanisms responsible for generalized spike-wave absence seizures are poorly understood. In mutant mice carrying loss-of-function (LOF) mutations in Cacna1a, which encodes the α1 pore-forming subunit of CaV 2.1 (P/Q-type) voltage-gated Ca(2+) channels, generalized spike-wave seizures have been suggested to result from excessive bursting of thalamocortical cells. However, other cellular populations including cortical inhibitory interneurons may contribute to this phenotype. We investigated how different cortical interneuron subtypes are affected by the loss of CaV 2.1 channel function and how this contributes to the onset of generalized epilepsy. METHODS We designed genetic strategies to induce a selective Cacna1a LOF mutation in different cortical γ-aminobutyric acidergic (GABAergic) and/or glutamatergic neuronal populations in mice. We assessed the cellular and network consequences of these mutations by combining immunohistochemical assays, in vitro physiology, optogenetics, and in vivo video electroencephalographic recordings. RESULTS We demonstrate that selective Cacna1a LOF from a subset of cortical interneurons, including parvalbumin (PV)(+) and somatostatin (SST)(+) interneurons, results in severe generalized epilepsy. Loss of CaV 2.1 channel function compromises GABA release from PV(+) but not SST(+) interneurons. Moreover, thalamocortical projection neurons do not show enhanced bursting in these mutants, suggesting that this feature is not essential for the development of generalized spike-wave seizures. Notably, the concurrent removal of CaV 2.1 channels in cortical pyramidal cells and interneurons considerably lessens seizure severity by decreasing cortical excitability. INTERPRETATION Our findings demonstrate that conditional ablation of CaV 2.1 channel function from cortical PV(+) interneurons alters GABA release from these cells, impairs their ability to constrain cortical pyramidal cell excitability, and is sufficient to cause generalized seizures.
Collapse
Affiliation(s)
- Elsa Rossignol
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY; Pediatric Neurology Department of Neuroscience, Saint Justine University Hospital Center, University of Montreal, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
48
|
Cheong E, Shin HS. T-type Ca²⁺ channels in absence epilepsy. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1560-71. [PMID: 23416255 DOI: 10.1016/j.bbamem.2013.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 01/15/2013] [Accepted: 02/01/2013] [Indexed: 11/28/2022]
Abstract
Low-voltage-activated T-type Ca²⁺ channels are highly expressed in the thalamocortical circuit, suggesting that they play a role in this brain circuit. Indeed, low-threshold burst firing mediated by T-type Ca²⁺ channels has long been implicated in the synchronization of the thalamocortical circuit. Over the past few decades, the conventional view has been that rhythmic burst firing mediated by T-type channels in both thalamic reticular nuclie (TRN) and thalamocortical (TC) neurons are equally critical in the generation of thalamocortical oscillations during sleep rhythms and spike-wave-discharges (SWDs). This review broadly investigates recent studies indicating that even though both TRN and TC nuclei are required for thalamocortical oscillations, the contributions of T-type channels to TRN and TC neurons are not equal in the genesis of sleep spindles and SWDs. T-type channels in TC neurons are an essential component of SWD generation, whereas the requirement for TRN T-type channels in SWD generation remains controversial at least in the GBL model of absence seizures. Therefore, a deeper understanding of the functional consequences of modulating each T-type channel subtype could guide the development of therapeutic tools for absence seizures while minimizing side effects on physiological thalamocortical oscillations. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- Eunji Cheong
- Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | | |
Collapse
|
49
|
Modulation of low-voltage-activated T-type Ca²⁺ channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1550-9. [PMID: 22975282 DOI: 10.1016/j.bbamem.2012.08.032] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/29/2012] [Accepted: 08/30/2012] [Indexed: 12/16/2022]
Abstract
Low-voltage-activated T-type Ca²⁺ channels contribute to a wide variety of physiological functions, most predominantly in the nervous, cardiovascular and endocrine systems. Studies have documented the roles of T-type channels in sleep, neuropathic pain, absence epilepsy, cell proliferation and cardiovascular function. Importantly, novel aspects of the modulation of T-type channels have been identified over the last few years, providing new insights into their physiological and pathophysiological roles. Although there is substantial literature regarding modulation of native T-type channels, the underlying molecular mechanisms have only recently begun to be addressed. This review focuses on recent evidence that the Ca(v)3 subunits of T-type channels, Ca(v)3.1, Ca(v)3.2 and Ca(v)3.3, are differentially modulated by a multitude of endogenous ligands including anandamide, monocyte chemoattractant protein-1, endostatin, and redox and oxidizing agents. The review also provides an overview of recent knowledge gained concerning downstream pathways involving G-protein-coupled receptors. This article is part of a Special Issue entitled: Calcium channels.
Collapse
|
50
|
Tringham E, Powell KL, Cain SM, Kuplast K, Mezeyova J, Weerapura M, Eduljee C, Jiang X, Smith P, Morrison JL, Jones NC, Braine E, Rind G, Fee-Maki M, Parker D, Pajouhesh H, Parmar M, O'Brien TJ, Snutch TP. T-type calcium channel blockers that attenuate thalamic burst firing and suppress absence seizures. Sci Transl Med 2012; 4:121ra19. [PMID: 22344687 DOI: 10.1126/scitranslmed.3003120] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Absence seizures are a common seizure type in children with genetic generalized epilepsy and are characterized by a temporary loss of awareness, arrest of physical activity, and accompanying spike-and-wave discharges on an electroencephalogram. They arise from abnormal, hypersynchronous neuronal firing in brain thalamocortical circuits. Currently available therapeutic agents are only partially effective and act on multiple molecular targets, including γ-aminobutyric acid (GABA) transaminase, sodium channels, and calcium (Ca(2+)) channels. We sought to develop high-affinity T-type specific Ca(2+) channel antagonists and to assess their efficacy against absence seizures in the Genetic Absence Epilepsy Rats from Strasbourg (GAERS) model. Using a rational drug design strategy that used knowledge from a previous N-type Ca(2+) channel pharmacophore and a high-throughput fluorometric Ca(2+) influx assay, we identified the T-type Ca(2+) channel blockers Z941 and Z944 as candidate agents and showed in thalamic slices that they attenuated burst firing of thalamic reticular nucleus neurons in GAERS. Upon administration to GAERS animals, Z941 and Z944 potently suppressed absence seizures by 85 to 90% via a mechanism distinct from the effects of ethosuximide and valproate, two first-line clinical drugs for absence seizures. The ability of the T-type Ca(2+) channel antagonists to inhibit absence seizures and to reduce the duration and cycle frequency of spike-and-wave discharges suggests that these agents have a unique mechanism of action on pathological thalamocortical oscillatory activity distinct from current drugs used in clinical practice.
Collapse
Affiliation(s)
- Elizabeth Tringham
- Zalicus Pharmaceuticals Ltd., Suite 301, 2389 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|