1
|
Cohen JD, You D, Sharma AK, Takai T, Hara H, Sales VT, Yukawa T, Cai B. In vitro human ion channel assays predictive of drug-induced seizure. Toxicol Sci 2025; 203:253-268. [PMID: 39661496 PMCID: PMC11775423 DOI: 10.1093/toxsci/kfae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Seizure is among the most severe FDA black box warnings of neurotoxicity reported on drug labels. Gaining a better mechanistic understanding of off-targets causative of seizure will improve the identification of potential seizure risks preclinically. In the present study, we evaluated an in vitro panel of 9 investigational (Cav2.1, Cav3.2, GlyRA1, AMPA, HCN1, Kv1.1, Kv7.2/7.3, NaV1.1, Nav1.2) and 2 standard (GABA-A, NMDA) ion channel targets with strong correlative links to seizure, using automated electrophysiology. Each target was assessed with a library of 34 preclinical compounds and 10 approved drugs with known effects of convulsion in vivo and/or in patients. Cav2.1 had the highest frequency of positive hits, 20 compounds with an EC30 or IC30 ≤ 30 µM, and the highest importance score relative to the 11 targets. An additional 35 approved drugs, with categorized low to frequent seizure risk in patients, were evaluated in the Cav2.1 assay. The Cav2.1 assay predicted preclinical compounds to cause convulsion in nonclinical species with a sensitivity of 52% and specificity of 78%, and approved drugs to cause seizure in nonclinical species or in patients with a sensitivity of 48% or 54% and specificity of 71% or 78%, respectively. The integrated panel of 11 ion channel targets predicted preclinical compounds to cause convulsion in nonclinical species with a sensitivity of 68%, specificity of 56%, and accuracy of 65%. This study highlights the utility of expanding the in vitro panel of targets evaluated for seizurogenic activity, in order to reduce compound attrition early on in drug discovery.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Dahea You
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Ashok K Sharma
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Takafumi Takai
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| | - Hideto Hara
- Drug Safety Research & Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Vicencia T Sales
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Tomoya Yukawa
- Drug Safety Research & Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Beibei Cai
- Drug Safety Research & Evaluation, Takeda Development Center Americas, Inc, San Diego, CA 92121-1964, United States
| |
Collapse
|
2
|
Pietrobon D, Brennan KC. Mechanisms underlying CSD initiation implicated by genetic mouse models of migraine. J Headache Pain 2025; 26:17. [PMID: 39871148 PMCID: PMC11773941 DOI: 10.1186/s10194-025-01948-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
A key unanswered question in migraine neurobiology concerns the mechanisms that make the brain of migraineurs susceptible to cortical spreading depression (CSD, a spreading depolarization that underlies migraine aura and may trigger the migraine pain mechanisms). Important insights into this question can be obtained by studying the mechanisms of facilitation of CSD initiation in genetic mouse models of the disease. These models, all generated from families with hereditary migraine, allow the investigation of the functional consequences of disease-causing mutations at the molecular, cellular, synaptic and neural circuit levels. In this review, after describing the available genetic mouse models of migraine, which all share increased susceptibility to experimentally induced CSD, we will discuss the functional alterations in their cerebral cortex and the mechanisms underlying the facilitation of CSD initiation in their cortex, as well as the insights that these mechanisms may give into the mechanisms of initiation of spontaneous CSDs in migraine.
Collapse
Affiliation(s)
- Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center, University of Padova, Via Ugo Bassi 58, 35131, Padua, Italy.
| | - K C Brennan
- Department of Neurology, University of Utah, 383 Colorow Drive, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
3
|
Griflyuk AV, Postnikova TY, Zaitsev AV. Animal Models of Febrile Seizures: Limitations and Recent Advances in the Field. Cells 2024; 13:1895. [PMID: 39594643 PMCID: PMC11592604 DOI: 10.3390/cells13221895] [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: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Febrile seizures (FSs) are defined as seizures occurring in children aged 6 months to 5 years with a background of elevated body temperature. It is one of the most common neurological disorders of childhood, emphasizing the importance of understanding the causes of FSs and their impact on the developing nervous system. However, there are significant limitations to the technologies currently available for studying the etiology and pathophysiology of seizures in humans. It is currently not possible to adequately capture the subtle molecular and structural rearrangements of the nervous system that can occur after seizures in humans. The use of animal models can be invaluable for these purposes. The most commonly used models in modern research are hyperthermic models in rats and mice aged 10-12 days. While these models can reproduce many of the characteristics of FSs, they have certain limitations. This review outlines the key considerations when working with models of FSs, provides an overview of current approaches to producing seizures in different model subjects, and presents a summary of key findings regarding morphological and functional changes in the brain and behavioral alterations that have been identified in studies using animal models of FSs.
Collapse
Affiliation(s)
| | | | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 44, Toreza Prospekt, Saint Petersburg 194223, Russia; (A.V.G.); (T.Y.P.)
| |
Collapse
|
4
|
Shore AN, Li K, Safari M, Qunies AM, Spitznagel BD, Weaver CD, Emmitte K, Frankel W, Weston MC. Heterozygous expression of a Kcnt1 gain-of-function variant has differential effects on somatostatin- and parvalbumin-expressing cortical GABAergic neurons. eLife 2024; 13:RP92915. [PMID: 39392867 PMCID: PMC11469685 DOI: 10.7554/elife.92915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024] Open
Abstract
More than 20 recurrent missense gain-of-function (GOF) mutations have been identified in the sodium-activated potassium (KNa) channel gene KCNT1 in patients with severe developmental and epileptic encephalopathies (DEEs), most of which are resistant to current therapies. Defining the neuron types most vulnerable to KCNT1 GOF will advance our understanding of disease mechanisms and provide refined targets for precision therapy efforts. Here, we assessed the effects of heterozygous expression of a Kcnt1 GOF variant (Kcnt1Y777H) on KNa currents and neuronal physiology among cortical glutamatergic and GABAergic neurons in mice, including those expressing vasoactive intestinal polypeptide (VIP), somatostatin (SST), and parvalbumin (PV), to identify and model the pathogenic mechanisms of autosomal dominant KCNT1 GOF variants in DEEs. Although the Kcnt1Y777H variant had no effects on glutamatergic or VIP neuron function, it increased subthreshold KNa currents in both SST and PV neurons but with opposite effects on neuronal output; SST neurons became hypoexcitable with a higher rheobase current and lower action potential (AP) firing frequency, whereas PV neurons became hyperexcitable with a lower rheobase current and higher AP firing frequency. Further neurophysiological and computational modeling experiments showed that the differential effects of the Kcnt1Y777H variant on SST and PV neurons are not likely due to inherent differences in these neuron types, but to an increased persistent sodium current in PV, but not SST, neurons. The Kcnt1Y777H variant also increased excitatory input onto, and chemical and electrical synaptic connectivity between, SST neurons. Together, these data suggest differential pathogenic mechanisms, both direct and compensatory, contribute to disease phenotypes, and provide a salient example of how a pathogenic ion channel variant can cause opposite functional effects in closely related neuron subtypes due to interactions with other ionic conductances.
Collapse
Affiliation(s)
- Amy N Shore
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology ResearchRoanokeUnited States
- Department of Neurological Sciences, University of VermontBurlingtonUnited States
| | - Keyong Li
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology ResearchRoanokeUnited States
| | - Mona Safari
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology ResearchRoanokeUnited States
- Translational Biology, Medicine, and Health Graduate Program, Fralin Biomedical Research Institute at Virginia Tech CarilionRoanokeUnited States
| | - Alshaima'a M Qunies
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science CenterFort WorthUnited States
- School of Biomedical Sciences, University of North Texas Health Science CenterFort WorthUnited States
| | - Brittany D Spitznagel
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashvilleUnited States
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
| | - C David Weaver
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashvilleUnited States
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
| | - Kyle Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science CenterFort WorthUnited States
| | - Wayne Frankel
- Institute for Genomic Medicine, Columbia UniversityNew YorkUnited States
- Department of Neurology, Columbia UniversityNew YorkUnited States
| | - Matthew C Weston
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology ResearchRoanokeUnited States
- Department of Neurological Sciences, University of VermontBurlingtonUnited States
- Translational Biology, Medicine, and Health Graduate Program, Fralin Biomedical Research Institute at Virginia Tech CarilionRoanokeUnited States
- School of Neuroscience, Virginia TechBlacksburgUnited States
| |
Collapse
|
5
|
Shore AN, Li K, Safari M, Qunies AM, Spitznagel BD, Weaver CD, Emmitte KA, Frankel WN, Weston MC. Heterozygous expression of a Kcnt1 gain-of-function variant has differential effects on SST- and PV-expressing cortical GABAergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.11.561953. [PMID: 37873369 PMCID: PMC10592778 DOI: 10.1101/2023.10.11.561953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
More than twenty recurrent missense gain-of-function (GOF) mutations have been identified in the sodium-activated potassium (KNa) channel gene KCNT1 in patients with severe developmental and epileptic encephalopathies (DEEs), most of which are resistant to current therapies. Defining the neuron types most vulnerable to KCNT1 GOF will advance our understanding of disease mechanisms and provide refined targets for precision therapy efforts. Here, we assessed the effects of heterozygous expression of a Kcnt1 GOF variant (Y777H) on KNa currents and neuronal physiology among cortical glutamatergic and GABAergic neurons in mice, including those expressing vasoactive intestinal polypeptide (VIP), somatostatin (SST), and parvalbumin (PV), to identify and model the pathogenic mechanisms of autosomal dominant KCNT1 GOF variants in DEEs. Although the Kcnt1-Y777H variant had no effects on glutamatergic or VIP neuron function, it increased subthreshold KNa currents in both SST and PV neurons but with opposite effects on neuronal output; SST neurons became hypoexcitable with a higher rheobase current and lower action potential (AP) firing frequency, whereas PV neurons became hyperexcitable with a lower rheobase current and higher AP firing frequency. Further neurophysiological and computational modeling experiments showed that the differential effects of the Y777H variant on SST and PV neurons are not likely due to inherent differences in these neuron types, but to an increased persistent sodium current in PV, but not SST, neurons. The Y777H variant also increased excitatory input onto, and chemical and electrical synaptic connectivity between, SST neurons. Together, these data suggest differential pathogenic mechanisms, both direct and compensatory, contribute to disease phenotypes, and provide a salient example of how a pathogenic ion channel variant can cause opposite functional effects in closely related neuron subtypes due to interactions with other ionic conductances.
Collapse
Affiliation(s)
- Amy N. Shore
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, VA, USA
- Department of Neurological Sciences, University of Vermont, Burlington, VT, USA
| | - Keyong Li
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, VA, USA
| | - Mona Safari
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, VA, USA
- Translational Biology, Medicine, and Health Graduate Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Alshaima’a M. Qunies
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
- School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Brittany D. Spitznagel
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Wayne N. Frankel
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Matthew C. Weston
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, VA, USA
- Department of Neurological Sciences, University of Vermont, Burlington, VT, USA
- Translational Biology, Medicine, and Health Graduate Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
6
|
Vinogradov A, Kapucu EF, Narkilahti S. Exploring Kainic Acid-Induced Alterations in Circular Tripartite Networks with Advanced Analysis Tools. eNeuro 2024; 11:ENEURO.0035-24.2024. [PMID: 39079743 PMCID: PMC11289587 DOI: 10.1523/eneuro.0035-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/26/2024] [Accepted: 06/10/2024] [Indexed: 08/02/2024] Open
Abstract
Brain activity implies the orchestrated functioning of interconnected brain regions. Typical in vitro models aim to mimic the brain using single human pluripotent stem cell-derived neuronal networks. However, the field is constantly evolving to model brain functions more accurately through the use of new paradigms, e.g., brain-on-a-chip models with compartmentalized structures and integrated sensors. These methods create novel data requiring more complex analysis approaches. The previously introduced circular tripartite network concept models the connectivity between spatially diverse neuronal structures. The model consists of a microfluidic device allowing axonal connectivity between separated neuronal networks with an embedded microelectrode array to record both local and global electrophysiological activity patterns in the closed circuitry. The existing tools are suboptimal for the analysis of the data produced with this model. Here, we introduce advanced tools for synchronization and functional connectivity assessment. We used our custom-designed analysis to assess the interrelations between the kainic acid (KA)-exposed proximal compartment and its nonexposed distal neighbors before and after KA. Novel multilevel circuitry bursting patterns were detected and analyzed in parallel with the inter- and intracompartmental functional connectivity. The effect of KA on the proximal compartment was captured, and the spread of this effect to the nonexposed distal compartments was revealed. KA induced divergent changes in bursting behaviors, which may be explained by distinct baseline activity and varied intra- and intercompartmental connectivity strengths. The circular tripartite network concept combined with our developed analysis advances importantly both face and construct validity in modeling human epilepsy in vitro.
Collapse
Affiliation(s)
- Andrey Vinogradov
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere 33520, Finland
| | - Emre Fikret Kapucu
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere 33520, Finland
| | - Susanna Narkilahti
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere 33520, Finland
| |
Collapse
|
7
|
Capitano F, Kuchenbuch M, Lavigne J, Chaptoukaev H, Zuluaga MA, Lorenzi M, Nabbout R, Mantegazza M. Preictal dysfunctions of inhibitory interneurons paradoxically lead to their rebound hyperactivity and to low-voltage-fast onset seizures in Dravet syndrome. Proc Natl Acad Sci U S A 2024; 121:e2316364121. [PMID: 38809712 PMCID: PMC11161744 DOI: 10.1073/pnas.2316364121] [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: 09/26/2023] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Epilepsies have numerous specific mechanisms. The understanding of neural dynamics leading to seizures is important for disclosing pathological mechanisms and developing therapeutic approaches. We investigated electrographic activities and neural dynamics leading to convulsive seizures in patients and mouse models of Dravet syndrome (DS), a developmental and epileptic encephalopathy in which hypoexcitability of GABAergic neurons is considered to be the main dysfunction. We analyzed EEGs from DS patients carrying a SCN1A pathogenic variant, as well as epidural electrocorticograms, hippocampal local field potentials, and hippocampal single-unit neuronal activities in Scn1a+/- and Scn1aRH/+ DS mice. Strikingly, most seizures had low-voltage-fast onset in both patients and mice, which is thought to be generated by hyperactivity of GABAergic interneurons, the opposite of the main pathological mechanism of DS. Analyzing single-unit recordings, we observed that temporal disorganization of the firing of putative interneurons in the period immediately before the seizure (preictal) precedes the increase of their activity at seizure onset, together with the entire neuronal network. Moreover, we found early signatures of the preictal period in the spectral features of hippocampal and cortical field potential of Scn1a mice and of patients' EEG, which are consistent with the dysfunctions that we observed in single neurons and that allowed seizure prediction. Therefore, the perturbed preictal activity of interneurons leads to their hyperactivity at the onset of generalized seizures, which have low-voltage-fast features that are similar to those observed in other epilepsies and are triggered by hyperactivity of GABAergic neurons. Preictal spectral features may be used as predictive seizure biomarkers.
Collapse
Affiliation(s)
- Fabrizio Capitano
- University Cote d’Azur, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis06560, France
- CNRS UMR 7275, Valbonne-Sophia Antipolis06560, France
- Inserm U1323, Valbonne-Sophia Antipolis06650, France
| | - Mathieu Kuchenbuch
- Reference Centre for Rare Epilepsies, Member of European Reference Network EpiCARE, Department of Pediatric Neurology, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris75015, France
- Laboratory of Translational Research for Neurological Disorders, Inserm UMR 1163, Imagine Institute, Université Paris Cité, Paris75015, France
| | - Jennifer Lavigne
- University Cote d’Azur, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis06560, France
- CNRS UMR 7275, Valbonne-Sophia Antipolis06560, France
- Inserm U1323, Valbonne-Sophia Antipolis06650, France
| | | | | | - Marco Lorenzi
- University Cote d’Azur, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis06560, France
- Epione Research team, Inria Center of Université Côte d’Azur, Biot-Sophia Antipolis06410, France
| | - Rima Nabbout
- Reference Centre for Rare Epilepsies, Member of European Reference Network EpiCARE, Department of Pediatric Neurology, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris75015, France
- Laboratory of Translational Research for Neurological Disorders, Inserm UMR 1163, Imagine Institute, Université Paris Cité, Paris75015, France
| | - Massimo Mantegazza
- University Cote d’Azur, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis06560, France
- CNRS UMR 7275, Valbonne-Sophia Antipolis06560, France
- Inserm U1323, Valbonne-Sophia Antipolis06650, France
| |
Collapse
|
8
|
Krüger J, Lerche H. Retigabine and gabapentin restore channel function and neuronal firing in a cellular model of an epilepsy-associated dominant-negative KCNQ5 variant. Neuropharmacology 2024; 250:109892. [PMID: 38428481 DOI: 10.1016/j.neuropharm.2024.109892] [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: 09/13/2023] [Revised: 01/19/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
KCNQ5 encodes the voltage-gated potassium channel KV7.5, a member of the KV7 channel family, which conducts the M-current. This current is a potent regulator of neuronal excitability by regulating membrane potential in the subthreshold range of action potentials and mediating the medium and slow afterhyperpolarization. Recently, we have identified five loss-of-function variants in KCNQ5 in patients with genetic generalized epilepsy. Using the most severe dominant-negative variant (R359C), we set out to investigate pharmacological therapeutic intervention by KV7 channel openers on channel function and neuronal firing. Retigabine and gabapentin increased R359C-derived M-current amplitudes in HEK cells expressing homomeric or heteromeric mutant KV7.5 channels. Retigabine was most effective in restoring K+ currents. Ten μM retigabine was sufficient to reach the level of WT currents without retigabine, whereas 100 μM of gabapentin showed less than half of this effect and application of 50 μM ZnCl2 only significantly increased M-current amplitude in heteromeric channels. Overexpression of KV7.5-WT potently inhibited neuronal firing by increasing the M-current, whereas R359C overexpression had the opposite effect and additionally decreased the medium afterhyperpolarization current. Both aforementioned drugs and Zn2+ reversed the effect of R359C expression by reducing firing to nearly normal levels at high current injections. Our study shows that a dominant-negative variant with a complete loss-of-function in KV7.5 leads to largely increased neuronal firing which may explain a neuronal hyperexcitability in patients. KV7 channel openers, such as retigabine or gabapentin, could be treatment options for patients currently displaying pharmacoresistant epilepsy and carrying loss-of-function variants in KCNQ5.
Collapse
Affiliation(s)
- Johanna Krüger
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Straße 27, 72076, Tübingen, Germany.
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Straße 27, 72076, Tübingen, Germany.
| |
Collapse
|
9
|
Wu J, Quraishi IH, Zhang Y, Bromwich M, Kaczmarek LK. Disease-causing Slack potassium channel mutations produce opposite effects on excitability of excitatory and inhibitory neurons. Cell Rep 2024; 43:113904. [PMID: 38457342 PMCID: PMC11013952 DOI: 10.1016/j.celrep.2024.113904] [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: 04/18/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/10/2024] Open
Abstract
The KCNT1 gene encodes the sodium-activated potassium channel Slack (KCNT1, KNa1.1), a regulator of neuronal excitability. Gain-of-function mutations in humans cause cortical network hyperexcitability, seizures, and severe intellectual disability. Using a mouse model expressing the Slack-R455H mutation, we find that Na+-dependent K+ (KNa) and voltage-dependent sodium (NaV) currents are increased in both excitatory and inhibitory cortical neurons. These increased currents, however, enhance the firing of excitability neurons but suppress that of inhibitory neurons. We further show that the expression of NaV channel subunits, particularly that of NaV1.6, is upregulated and that the length of the axon initial segment and of axonal NaV immunostaining is increased in both neuron types. Our study on the coordinate regulation of KNa currents and the expression of NaV channels may provide an avenue for understanding and treating epilepsies and other neurological disorders.
Collapse
Affiliation(s)
- Jing Wu
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Imran H Quraishi
- Department of Neurology, Yale Comprehensive Epilepsy Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mark Bromwich
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
10
|
Peall KJ, Owen MJ, Hall J. Rare genetic brain disorders with overlapping neurological and psychiatric phenotypes. Nat Rev Neurol 2024; 20:7-21. [PMID: 38001363 DOI: 10.1038/s41582-023-00896-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 11/26/2023]
Abstract
Understanding rare genetic brain disorders with overlapping neurological and psychiatric phenotypes is of increasing importance given the potential for developing disease models that could help to understand more common, polygenic disorders. However, the traditional clinical boundaries between neurology and psychiatry result in frequent segregation of these disorders into distinct silos, limiting cross-specialty understanding that could facilitate clinical and biological advances. In this Review, we highlight multiple genetic brain disorders in which neurological and psychiatric phenotypes are observed, but for which in-depth, cross-spectrum clinical phenotyping is rarely undertaken. We describe the combined phenotypes observed in association with genetic variants linked to epilepsy, dystonia, autism spectrum disorder and schizophrenia. We also consider common underlying mechanisms that centre on synaptic plasticity, including changes to synaptic and neuronal structure, calcium handling and the balance of excitatory and inhibitory neuronal activity. Further investigation is needed to better define and replicate these phenotypes in larger cohorts, which would help to gain greater understanding of the pathophysiological mechanisms and identify common therapeutic targets.
Collapse
Affiliation(s)
- Kathryn J Peall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK.
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Michael J Owen
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
11
|
Smith J, Richerson G, Kouchi H, Duprat F, Mantegazza M, Bezin L, Rheims S. Are we there yet? A critical evaluation of sudden and unexpected death in epilepsy models. Epilepsia 2024; 65:9-25. [PMID: 37914406 DOI: 10.1111/epi.17812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 11/03/2023]
Abstract
Although animal models have helped to elaborate meaningful hypotheses about the pathophysiology of sudden and unexpected death in epilepsy (SUDEP), specific prevention strategies are still lacking, potentially reflecting the limitations of these models and the intrinsic difficulties of investigating SUDEP. The interpretation of preclinical data and their translation to diagnostic and therapeutic developments in patients thus require a high level of confidence in their relevance to model the human situation. Preclinical models of SUDEP are heterogeneous and include rodent and nonrodent species. A critical aspect is whether the animals have isolated seizures exclusively induced by a specific trigger, such as models where seizures are elicited by electrical stimulation, pharmacological intervention, or DBA mouse strains, or whether they suffer from epilepsy with spontaneous seizures, with or without spontaneous SUDEP, either of nongenetic epilepsy etiology or from genetically based developmental and epileptic encephalopathies. All these models have advantages and potential disadvantages, but it is important to be aware of these limitations to interpret data appropriately in a translational perspective. The majority of models with spontaneous seizures are of a genetic basis, whereas SUDEP cases with a genetic basis represent only a small proportion of the total number. In almost all models, cardiorespiratory arrest occurs during the course of the seizure, contrary to that in patients observed at the time of death, potentially raising the issue of whether we are studying models of SUDEP or models of periseizure death. However, some of these limitations are impossible to avoid and can in part be dependent on specific features of SUDEP, which may be difficult to model. Several preclinical tools are available to address certain gaps in SUDEP pathophysiology, which can be used to further validate current preclinical models.
Collapse
Affiliation(s)
- Jonathon Smith
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
| | - George Richerson
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Hayet Kouchi
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
| | - Fabrice Duprat
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
| | - Massimo Mantegazza
- University Cote d'Azur, Valbonne-Sophia Antipolis, France
- CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
| | - Laurent Bezin
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
| | - Sylvain Rheims
- Lyon Neuroscience Research Center (CRNL, INSERM U1028/CNRS UMR 5292, Lyon 1 University), Lyon, France
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and Lyon 1 University, Lyon, France
| |
Collapse
|
12
|
Lyu H, Boßelmann CM, Johannesen KM, Koko M, Ortigoza-Escobar JD, Aguilera-Albesa S, Garcia-Navas Núñez D, Linnankivi T, Gaily E, van Ruiten HJA, Richardson R, Betzler C, Horvath G, Brilstra E, Geerdink N, Orsucci D, Tessa A, Gardella E, Fleszar Z, Schöls L, Lerche H, Møller RS, Liu Y. Clinical and electrophysiological features of SCN8A variants causing episodic or chronic ataxia. EBioMedicine 2023; 98:104855. [PMID: 38251463 PMCID: PMC10628346 DOI: 10.1016/j.ebiom.2023.104855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Variants in SCN8A are associated with a spectrum of epilepsies and neurodevelopmental disorders. Ataxia as a predominant symptom of SCN8A variation has not been well studied. We set out to investigate disease mechanisms and genotype-phenotype correlations of SCN8A-related ataxia. METHODS We collected genetic and electro-clinical data of ten individuals from nine unrelated families carrying novel SCN8A variants associated with chronic progressive or episodic ataxia. Electrophysiological characterizations of these variants were performed in ND7/23 cells and cultured neurons. FINDINGS Variants associated with chronic progressive ataxia either decreased Na+ current densities and shifted activation curves towards more depolarized potentials (p.Asn995Asp, p.Lys1498Glu and p.Trp1266Cys) or resulted in a premature stop codon (p.Trp937Ter). Three variants (p.Arg847Gln and biallelic p.Arg191Trp/p.Asp1525Tyr) were associated with episodic ataxia causing loss-of-function by decreasing Na+ current densities or a hyperpolarizing shift of the inactivation curve. Two additional episodic ataxia-associated variants caused mixed gain- and loss-of function effects in ND7/23 cells and were further examined in primary murine hippocampal neuronal cultures. Neuronal firing in excitatory neurons was increased by p.Arg1629His, but decreased by p.Glu1201Lys. Neuronal firing in inhibitory neurons was decreased for both variants. No functional effect was observed for p.Arg1913Trp. In four individuals, treatment with sodium channel blockers exacerbated symptoms. INTERPRETATION We identified episodic or chronic ataxia as predominant phenotypes caused by variants in SCN8A. Genotype-phenotype correlations revealed a more pronounced loss-of-function effect for variants causing chronic ataxia. Sodium channel blockers should be avoided under these conditions. FUNDING BMBF, DFG, the Italian Ministry of Health, University of Tuebingen.
Collapse
Affiliation(s)
- Hang Lyu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Christian M Boßelmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Katrine M Johannesen
- Department of Clinical Genetics, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark; Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark
| | - Mahmoud Koko
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Juan Dario Ortigoza-Escobar
- Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, CIBERER-ISCIII and European Reference Network for Rare Neurological Diseases (ERN-RND), Barcelona, Spain
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitario de Navarra, Pamplona, Spain; Navarrabiomed-Fundación Miguel Servet, Pamplona, Spain
| | | | - Tarja Linnankivi
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Eija Gaily
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Henriette J A van Ruiten
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Ruth Richardson
- Northern Genetics Service, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, UK
| | - Cornelia Betzler
- Institute for Rehabilitation, Transition and Palliation, Paracelsus Medical University, Salzburg, Austria; Specialist Center for Paediatric Neurology, Neuro-Rehabilitation and Epileptology, Schön Klinik Vogtareuth, Germany
| | - Gabriella Horvath
- Adult Metabolic Diseases Clinic, BC Children's Hospital, Vancouver, Canada
| | - Eva Brilstra
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niels Geerdink
- Department of Pediatrics, Rijnstate Hospital, Arnhem, the Netherlands
| | | | | | - Elena Gardella
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark; Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Zofia Fleszar
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ludger Schöls
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark; Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Yuanyuan Liu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
13
|
Studtmann C, Ladislav M, Safari M, Khondaker R, Chen Y, Vaughan GA, Topolski MA, Tomović E, Balík A, Swanger SA. Ventral posterolateral and ventral posteromedial thalamocortical neurons have distinct physiological properties. J Neurophysiol 2023; 130:1492-1507. [PMID: 37937368 PMCID: PMC11068404 DOI: 10.1152/jn.00525.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/09/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023] Open
Abstract
Somatosensory information is propagated from the periphery to the cerebral cortex by two parallel pathways through the ventral posterolateral (VPL) and ventral posteromedial (VPM) thalamus. VPL and VPM neurons receive somatosensory signals from the body and head, respectively. VPL and VPM neurons may also receive cell type-specific GABAergic input from the reticular nucleus of the thalamus. Although VPL and VPM neurons have distinct connectivity and physiological roles, differences in their functional properties remain unclear as they are often studied as one ventrobasal thalamus neuron population. Here, we directly compared synaptic and intrinsic properties of VPL and VPM neurons in C57Bl/6J mice of both sexes aged P25-P32. VPL neurons showed greater depolarization-induced spike firing and spike frequency adaptation than VPM neurons. VPL and VPM neurons fired similar numbers of spikes during hyperpolarization rebound bursts, but VPM neurons exhibited shorter burst latency compared with VPL neurons, which correlated with larger sag potential. VPM neurons had larger membrane capacitance and more complex dendritic arbors. Recordings of spontaneous and evoked synaptic transmission suggested that VPL neurons receive stronger excitatory synaptic input, whereas inhibitory synapse strength was stronger in VPM neurons. This work indicates that VPL and VPM thalamocortical neurons have distinct intrinsic and synaptic properties. The observed functional differences could have important implications for their specific physiological and pathophysiological roles within the somatosensory thalamocortical network.NEW & NOTEWORTHY This study revealed that somatosensory thalamocortical neurons in the VPL and VPM have substantial differences in excitatory synaptic input and intrinsic firing properties. The distinct properties suggest that VPL and VPM neurons could process somatosensory information differently and have selective vulnerability to disease. This work improves our understanding of nucleus-specific neuron function in the thalamus and demonstrates the critical importance of studying these parallel somatosensory pathways separately.
Collapse
Affiliation(s)
- Carleigh Studtmann
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Marek Ladislav
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
| | - Mona Safari
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Rabeya Khondaker
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Yang Chen
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Grace A Vaughan
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, United States
| | - Mackenzie A Topolski
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
| | - Eni Tomović
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Balík
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Sharon A Swanger
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| |
Collapse
|
14
|
Koch NA, Sonnenberg L, Hedrich UBS, Lauxmann S, Benda J. Loss or gain of function? Effects of ion channel mutations on neuronal firing depend on the neuron type. Front Neurol 2023; 14:1194811. [PMID: 37292138 PMCID: PMC10244640 DOI: 10.3389/fneur.2023.1194811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Clinically relevant mutations to voltage-gated ion channels, called channelopathies, alter ion channel function, properties of ionic currents, and neuronal firing. The effects of ion channel mutations are routinely assessed and characterized as loss of function (LOF) or gain of function (GOF) at the level of ionic currents. However, emerging personalized medicine approaches based on LOF/GOF characterization have limited therapeutic success. Potential reasons are among others that the translation from this binary characterization to neuronal firing is currently not well-understood-especially when considering different neuronal cell types. In this study, we investigate the impact of neuronal cell type on the firing outcome of ion channel mutations. Methods To this end, we simulated a diverse collection of single-compartment, conductance-based neuron models that differed in their composition of ionic currents. We systematically analyzed the effects of changes in ion current properties on firing in different neuronal types. Additionally, we simulated the effects of known mutations in KCNA1 gene encoding the KV1.1 potassium channel subtype associated with episodic ataxia type 1 (EA1). Results These simulations revealed that the outcome of a given change in ion channel properties on neuronal excitability depends on neuron type, i.e., the properties and expression levels of the unaffected ionic currents. Discussion Consequently, neuron-type specific effects are vital to a full understanding of the effects of channelopathies on neuronal excitability and are an important step toward improving the efficacy and precision of personalized medicine approaches.
Collapse
Affiliation(s)
- Nils A. Koch
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Bernstein Center for Computational Neuroscience Tübingen, Tübingen, Germany
| | - Lukas Sonnenberg
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Bernstein Center for Computational Neuroscience Tübingen, Tübingen, Germany
| | - Ulrike B. S. Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stephan Lauxmann
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jan Benda
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Bernstein Center for Computational Neuroscience Tübingen, Tübingen, Germany
| |
Collapse
|
15
|
Bayraktar E, Liu Y, Sonnenberg L, Hedrich UBS, Sara Y, Eltokhi A, Lyu H, Lerche H, Wuttke TV, Lauxmann S. In vitro effects of eslicarbazepine (S-licarbazepine) as a potential precision therapy on SCN8A variants causing neuropsychiatric disorders. Br J Pharmacol 2023; 180:1038-1055. [PMID: 36321697 DOI: 10.1111/bph.15981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Variants in SCN8A, the NaV 1.6 channel's coding gene, are characterized by a variety of symptoms, including intractable epileptic seizures, psychomotor delay, progressive cognitive decline, autistic features, ataxia or dystonia. Standard anticonvulsant treatment has a limited impact on the course of disease. EXPERIMENTAL APPROACH We investigated the therapeutic potential of eslicarbazepine (S-licarbazepine; S-lic), an enhancer of slow inactivation of voltage gated sodium channels, on two variants with biophysical and neuronal gain-of-function (G1475R and M1760I) and one variant with biophysical gain-of-function but neuronal loss-of-function (A1622D) in neuroblastoma cells and in murine primary hippocampal neuron cultures. These three variants cover the broad spectrum of NaV 1.6-associated disease and are linked to representative phenotypes of mild to moderate epilepsy (G1475R), developmental and epileptic encephalopathy (M1760I) and intellectual disability without epilepsy (A1622D). KEY RESULTS Similar to known effects on NaV 1.6 wildtype channels, S-lic predominantly enhances slow inactivation on all tested variants, irrespective of their particular biophysical mechanisms. Beyond that, S-lic exhibits variant-specific effects including a partial reversal of pathologically slowed fast inactivation dynamics (A1622D and M1760I) and a trend to reduce enhanced persistent Na+ current by A1622D variant channels. Furthermore, our data in primary transfected neurons reveal that not only variant-associated hyperexcitability (M1760I and G1475R) but also hypoexcitability (A1622D) can be modulated by S-lic. CONCLUSIONS AND IMPLICATIONS S-lic has not only substance-specific effects but also variant-specific effects. Personalized treatment regimens optimized to achieve such variant-specific pharmacological modulation may help to reduce adverse side effects and improve the overall therapeutic outcome of SCN8A-related disease.
Collapse
Affiliation(s)
- Erva Bayraktar
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Medical Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Yuanyuan Liu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Sonnenberg
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Neurobiology, University of Tübingen, Tübingen, Germany
| | - Ulrike B S Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Yildirim Sara
- Department of Medical Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ahmed Eltokhi
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | - Hang Lyu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Stephan Lauxmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Conti F, Pietrobon D. Astrocytic Glutamate Transporters and Migraine. Neurochem Res 2023; 48:1167-1179. [PMID: 36583835 DOI: 10.1007/s11064-022-03849-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/31/2022]
Abstract
Glutamate levels and lifetime in the brain extracellular space are dinamically regulated by a family of Na+- and K+-dependent glutamate transporters, which thereby control numerous brain functions and play a role in numerous neurological and psychiatric diseases. Migraine is a neurological disorder characterized by recurrent attacks of typically throbbing and unilateral headache and by a global dysfunction in multisensory processing. Familial hemiplegic migraine type 2 (FHM2) is a rare monogenic form of migraine with aura caused by loss-of-function mutations in the α2 Na/K ATPase (α2NKA). In the adult brain, this pump is expressed almost exclusively in astrocytes where it is colocalized with glutamate transporters. Knockin mouse models of FHM2 (FHM2 mice) show a reduced density of glutamate transporters in perisynaptic astrocytic processes (mirroring the reduced expression of α2NKA) and a reduced rate of glutamate clearance at cortical synapses during neuronal activity and sensory stimulation. Here we review the migraine-relevant alterations produced by the astrocytic glutamate transport dysfunction in FHM2 mice and their underlying mechanisms, in particular regarding the enhanced brain susceptibility to cortical spreading depression (the phenomenon that underlies migraine aura and can also initiate the headache mechanisms) and the enhanced algesic response to a migraine trigger.
Collapse
Affiliation(s)
- Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.
| | - Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center (PNC), University of Padova, 35131, Padua, Italy.
- CNR Institute of Neuroscience, 35131, Padua, Italy.
| |
Collapse
|
17
|
Bauer J, Devinsky O, Rothermel M, Koch H. Autonomic dysfunction in epilepsy mouse models with implications for SUDEP research. Front Neurol 2023; 13:1040648. [PMID: 36686527 PMCID: PMC9853197 DOI: 10.3389/fneur.2022.1040648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
Epilepsy has a high prevalence and can severely impair quality of life and increase the risk of premature death. Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death in drug-resistant epilepsy and most often results from respiratory and cardiac impairments due to brainstem dysfunction. Epileptic activity can spread widely, influencing neuronal activity in regions outside the epileptic network. The brainstem controls cardiorespiratory activity and arousal and reciprocally connects to cortical, diencephalic, and spinal cord areas. Epileptic activity can propagate trans-synaptically or via spreading depression (SD) to alter brainstem functions and cause cardiorespiratory dysfunction. The mechanisms by which seizures propagate to or otherwise impair brainstem function and trigger the cascading effects that cause SUDEP are poorly understood. We review insights from mouse models combined with new techniques to understand the pathophysiology of epilepsy and SUDEP. These techniques include in vivo, ex vivo, invasive and non-invasive methods in anesthetized and awake mice. Optogenetics combined with electrophysiological and optical manipulation and recording methods offer unique opportunities to study neuronal mechanisms under normal conditions, during and after non-fatal seizures, and in SUDEP. These combined approaches can advance our understanding of brainstem pathophysiology associated with seizures and SUDEP and may suggest strategies to prevent SUDEP.
Collapse
Affiliation(s)
- Jennifer Bauer
- Department of Epileptology and Neurology, RWTH Aachen University, Aachen, Germany,Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Orrin Devinsky
- Departments of Neurology, Neurosurgery and Psychiatry, NYU Langone School of Medicine, New York, NY, United States
| | - Markus Rothermel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Henner Koch
- Department of Epileptology and Neurology, RWTH Aachen University, Aachen, Germany,*Correspondence: Henner Koch ✉
| |
Collapse
|
18
|
Bryson A, Petrou S. SCN1A channelopathies: Navigating from genotype to neural circuit dysfunction. Front Neurol 2023; 14:1173460. [PMID: 37139072 PMCID: PMC10149698 DOI: 10.3389/fneur.2023.1173460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
The SCN1A gene is strongly associated with epilepsy and plays a central role for supporting cortical excitation-inhibition balance through the expression of NaV1.1 within inhibitory interneurons. The phenotype of SCN1A disorders has been conceptualized as driven primarily by impaired interneuron function that predisposes to disinhibition and cortical hyperexcitability. However, recent studies have identified SCN1A gain-of-function variants associated with epilepsy, and the presence of cellular and synaptic changes in mouse models that point toward homeostatic adaptations and complex network remodeling. These findings highlight the need to understand microcircuit-scale dysfunction in SCN1A disorders to contextualize genetic and cellular disease mechanisms. Targeting the restoration of microcircuit properties may be a fruitful strategy for the development of novel therapies.
Collapse
Affiliation(s)
- Alexander Bryson
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Alexander Bryson,
| | - Steven Petrou
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Praxis Precision Medicines, Inc., Cambridge, MA, United States
| |
Collapse
|
19
|
Brunklaus A, Brünger T, Feng T, Fons C, Lehikoinen A, Panagiotakaki E, Vintan MA, Symonds J, Andrew J, Arzimanoglou A, Delima S, Gallois J, Hanrahan D, Lesca G, MacLeod S, Marjanovic D, McTague A, Nuñez-Enamorado N, Perez-Palma E, Scott Perry M, Pysden K, Russ-Hall SJ, Scheffer IE, Sully K, Syrbe S, Vaher U, Velayutham M, Vogt J, Weiss S, Wirrell E, Zuberi SM, Lal D, Møller RS, Mantegazza M, Cestèle S. The gain of function SCN1A disorder spectrum: novel epilepsy phenotypes and therapeutic implications. Brain 2022; 145:3816-3831. [PMID: 35696452 PMCID: PMC9679167 DOI: 10.1093/brain/awac210] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/14/2022] [Accepted: 05/26/2022] [Indexed: 11/29/2022] Open
Abstract
Brain voltage-gated sodium channel NaV1.1 (SCN1A) loss-of-function variants cause the severe epilepsy Dravet syndrome, as well as milder phenotypes associated with genetic epilepsy with febrile seizures plus. Gain of function SCN1A variants are associated with familial hemiplegic migraine type 3. Novel SCN1A-related phenotypes have been described including early infantile developmental and epileptic encephalopathy with movement disorder, and more recently neonatal presentations with arthrogryposis. Here we describe the clinical, genetic and functional evaluation of affected individuals. Thirty-five patients were ascertained via an international collaborative network using a structured clinical questionnaire and from the literature. We performed whole-cell voltage-clamp electrophysiological recordings comparing sodium channels containing wild-type versus variant NaV1.1 subunits. Findings were related to Dravet syndrome and familial hemiplegic migraine type 3 variants. We identified three distinct clinical presentations differing by age at onset and presence of arthrogryposis and/or movement disorder. The most severely affected infants (n = 13) presented with congenital arthrogryposis, neonatal onset epilepsy in the first 3 days of life, tonic seizures and apnoeas, accompanied by a significant movement disorder and profound intellectual disability. Twenty-one patients presented later, between 2 weeks and 3 months of age, with a severe early infantile developmental and epileptic encephalopathy and a movement disorder. One patient presented after 3 months with developmental and epileptic encephalopathy only. Associated SCN1A variants cluster in regions of channel inactivation associated with gain of function, different to Dravet syndrome variants (odds ratio = 17.8; confidence interval = 5.4-69.3; P = 1.3 × 10-7). Functional studies of both epilepsy and familial hemiplegic migraine type 3 variants reveal alterations of gating properties in keeping with neuronal hyperexcitability. While epilepsy variants result in a moderate increase in action current amplitude consistent with mild gain of function, familial hemiplegic migraine type 3 variants induce a larger effect on gating properties, in particular the increase of persistent current, resulting in a large increase of action current amplitude, consistent with stronger gain of function. Clinically, 13 out of 16 (81%) gain of function variants were associated with a reduction in seizures in response to sodium channel blocker treatment (carbamazepine, oxcarbazepine, phenytoin, lamotrigine or lacosamide) without evidence of symptom exacerbation. Our study expands the spectrum of gain of function SCN1A-related epilepsy phenotypes, defines key clinical features, provides novel insights into the underlying disease mechanisms between SCN1A-related epilepsy and familial hemiplegic migraine type 3, and identifies sodium channel blockers as potentially efficacious therapies. Gain of function disease should be considered in early onset epilepsies with a pathogenic SCN1A variant and non-Dravet syndrome phenotype.
Collapse
Affiliation(s)
- Andreas Brunklaus
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Member of the ERN EpiCARE, Glasgow, UK
| | - Tobias Brünger
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Tony Feng
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Member of the ERN EpiCARE, Glasgow, UK
| | - Carmen Fons
- Pediatric Neurology Department, CIBERER-ISCIII, Sant Joan de Déu Universitary Hospital, Institut de Recerca Sant Joan de Déu, Member of the ERN EpiCARE, Barcelona, Spain
| | - Anni Lehikoinen
- Pediatric Neurology Department, Kuopio University Hospital, Member of the ERN EpiCARE, Kuopio, Finland
| | - Eleni Panagiotakaki
- Department of Paediatric Clinical Epileptology, sleep disorders and functional neurology, Member of the ERN EpiCARE, University Hospitals of Lyon (HCL) and Inserm U1028/CNRS UMR5292, Lyon, France
| | - Mihaela-Adela Vintan
- ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Neurology and Pediatric Neurology, Victor Babes, 43, 400012 Cluj-Napoca, Romania
| | - Joseph Symonds
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Member of the ERN EpiCARE, Glasgow, UK
| | - James Andrew
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Member of the ERN EpiCARE, Glasgow, UK
| | - Alexis Arzimanoglou
- Pediatric Neurology Department, CIBERER-ISCIII, Sant Joan de Déu Universitary Hospital, Institut de Recerca Sant Joan de Déu, Member of the ERN EpiCARE, Barcelona, Spain
- Department of Paediatric Clinical Epileptology, sleep disorders and functional neurology, Member of the ERN EpiCARE, University Hospitals of Lyon (HCL) and Inserm U1028/CNRS UMR5292, Lyon, France
| | - Sarah Delima
- Indiana University School of Medicine, IU Health Riley Hospital for Children, Department of Neurology, Division of Pediatric Neurology, Indianapolis, IN, USA
| | - Julie Gallois
- Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA, USA
| | - Donncha Hanrahan
- Department of Paediatric Neurology, Royal Belfast Hospital for Sick Children, Belfast, UK
| | - Gaetan Lesca
- Department of Medical Genetics, Lyon University Hospital, Member of the ERN EpiCARE, Université Claude Bernard Lyon 1, Lyon, France
| | - Stewart MacLeod
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Member of the ERN EpiCARE, Glasgow, UK
| | - Dragan Marjanovic
- The Danish Epilepsy Centre, Member of the ERN EpiCARE, Dianalund, Denmark
| | - Amy McTague
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Neurology, Great Ormond Street Hospital for Children, Member of the ERN EpiCARE, London, UK
| | | | - Eduardo Perez-Palma
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Santiago, Chile
| | - M Scott Perry
- Jane and John Justin Neurosciences Center, Cook Children’s Medical Center, Ft Worth, TX, USA
| | - Karen Pysden
- Paediatric Neurology Department, Leeds Teaching Hospitals, Leeds General Infirmary, Leeds, UK
| | - Sophie J Russ-Hall
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
- Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- Murdoch Children’s Research Institute and Department of Paediatrics, University of Melbourne, Royal Children’s Hospital, Melbourne, Australia
| | - Krystal Sully
- Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital, Houston, TX, USA
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Ulvi Vaher
- Children’s Clinic of Tartu University Hospital, Faculty of Medicine of Tartu University, Member of the ERN EpiCARE, Tartu, Estonia
| | | | - Julie Vogt
- West Midlands Regional Genetics Service, Birmingham Women’s and Children’s Hospital, Birmingham, UK
| | - Shelly Weiss
- Division of Neurology, SickKids, University of Toronto, Toronto, Canada
| | - Elaine Wirrell
- Divisions of Epilepsy and Child and Adolescent Neurology, Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Sameer M Zuberi
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Member of the ERN EpiCARE, Glasgow, UK
| | - Dennis Lal
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Stanley Center for Psychiatric Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rikke S Møller
- The Danish Epilepsy Centre, Member of the ERN EpiCARE, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Massimo Mantegazza
- Université Côte d’Azur, 06560 Valbonne-Sophia Antipolis, France
- CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), 06560 Valbonne-Sophia Antipolis, France
- Inserm, 06560 Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Côte d’Azur, 06560 Valbonne-Sophia Antipolis, France
- CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), 06560 Valbonne-Sophia Antipolis, France
| |
Collapse
|
20
|
Ye H, Hendee J, Ruan J, Zhirova A, Ye J, Dima M. Neuron matters: neuromodulation with electromagnetic stimulation must consider neurons as dynamic identities. J Neuroeng Rehabil 2022; 19:116. [PMID: 36329492 PMCID: PMC9632094 DOI: 10.1186/s12984-022-01094-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/15/2022] [Indexed: 11/06/2022] Open
Abstract
Neuromodulation with electromagnetic stimulation is widely used for the control of abnormal neural activity, and has been proven to be a valuable alternative to pharmacological tools for the treatment of many neurological diseases. Tremendous efforts have been focused on the design of the stimulation apparatus (i.e., electrodes and magnetic coils) that delivers the electric current to the neural tissue, and the optimization of the stimulation parameters. Less attention has been given to the complicated, dynamic properties of the neurons, and their context-dependent impact on the stimulation effects. This review focuses on the neuronal factors that influence the outcomes of electromagnetic stimulation in neuromodulation. Evidence from multiple levels (tissue, cellular, and single ion channel) are reviewed. Properties of the neural elements and their dynamic changes play a significant role in the outcome of electromagnetic stimulation. This angle of understanding yields a comprehensive perspective of neural activity during electrical neuromodulation, and provides insights in the design and development of novel stimulation technology.
Collapse
Affiliation(s)
- Hui Ye
- grid.164971.c0000 0001 1089 6558Department of Biology, Quinlan Life Sciences Education and Research Center, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660 USA
| | - Jenna Hendee
- grid.164971.c0000 0001 1089 6558Department of Biology, Quinlan Life Sciences Education and Research Center, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660 USA
| | - Joyce Ruan
- grid.164971.c0000 0001 1089 6558Department of Biology, Quinlan Life Sciences Education and Research Center, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660 USA
| | - Alena Zhirova
- grid.164971.c0000 0001 1089 6558Department of Biology, Quinlan Life Sciences Education and Research Center, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660 USA
| | - Jayden Ye
- grid.164971.c0000 0001 1089 6558Department of Biology, Quinlan Life Sciences Education and Research Center, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660 USA
| | - Maria Dima
- grid.164971.c0000 0001 1089 6558Department of Biology, Quinlan Life Sciences Education and Research Center, Loyola University Chicago, 1032 W. Sheridan Rd., Chicago, IL 60660 USA
| |
Collapse
|
21
|
Merseburg A, Kasemir J, Buss EW, Leroy F, Bock T, Porro A, Barnett A, Tröder SE, Engeland B, Stockebrand M, Moroni A, Siegelbaum S, Isbrandt D, Santoro B. Seizures, behavioral deficits and adverse drug responses in two new genetic mouse models of HCN1 epileptic encephalopathy. eLife 2022; 11:70826. [PMID: 35972069 PMCID: PMC9481245 DOI: 10.7554/elife.70826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
De novo mutations in voltage- and ligand-gated channels have been associated with an increasing number of cases of developmental and epileptic encephalopathies, which often fail to respond to classic antiseizure medications. Here, we examine two knock-in mouse models replicating de novo sequence variations in the HCN1 voltage-gated channel gene, p.G391D and p.M153I (Hcn1G380D/+ and Hcn1M142I/+ in mouse), associated with severe drug-resistant neonatal- and childhood-onset epilepsy, respectively. Heterozygous mice from both lines displayed spontaneous generalized tonic-clonic seizures. Animals replicating the p.G391D variant had an overall more severe phenotype, with pronounced alterations in the levels and distribution of HCN1 protein, including disrupted targeting to the axon terminals of basket cell interneurons. In line with clinical reports from patients with pathogenic HCN1 sequence variations, administration of the antiepileptic Na+ channel antagonists lamotrigine and phenytoin resulted in the paradoxical induction of seizures in both mouse lines, consistent with an effect to further impair inhibitory neuron function. We also show that these variants can render HCN1 channels unresponsive to classic antagonists, indicating the need to screen mutated channels to identify novel compounds with diverse mechanism of action. Our results underscore the necessity of tailoring effective therapies for specific channel gene variants, and how strongly validated animal models may provide an invaluable tool towards reaching this objective.
Collapse
Affiliation(s)
- Andrea Merseburg
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Jacquelin Kasemir
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Eric W Buss
- Department of Neuroscience, Columbia University, New York, United States
| | - Felix Leroy
- Department of Neuroscience, Columbia University, New York, United States
| | - Tobias Bock
- Department of Neuroscience, Columbia University, New York, United States
| | | | - Anastasia Barnett
- Department of Neuroscience, Columbia University, New York, United States
| | - Simon E Tröder
- Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Birgit Engeland
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Malte Stockebrand
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy
| | - Steve Siegelbaum
- Department of Neuroscience, Columbia University, New York, United States
| | - Dirk Isbrandt
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Cologne, Germany
| | - Bina Santoro
- Department of Neuroscience, Columbia University, New York, United States
| |
Collapse
|
22
|
Scalise S, Zannino C, Lucchino V, Lo Conte M, Scaramuzzino L, Cifelli P, D’Andrea T, Martinello K, Fucile S, Palma E, Gambardella A, Ruffolo G, Cuda G, Parrotta EI. Human iPSC Modeling of Genetic Febrile Seizure Reveals Aberrant Molecular and Physiological Features Underlying an Impaired Neuronal Activity. Biomedicines 2022; 10:biomedicines10051075. [PMID: 35625812 PMCID: PMC9138645 DOI: 10.3390/biomedicines10051075] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Mutations in SCN1A gene, encoding the voltage-gated sodium channel (VGSC) NaV1.1, are widely recognized as a leading cause of genetic febrile seizures (FS), due to the decrease in the Na+ current density, mainly affecting the inhibitory neuronal transmission. Here, we generated induced pluripotent stem cells (iPSCs)-derived neurons (idNs) from a patient belonging to a genetically well-characterized Italian family, carrying the c.434T > C mutation in SCN1A gene (hereafter SCN1AM145T). A side-by-side comparison of diseased and healthy idNs revealed an overall maturation delay of SCN1AM145T cells. Membranes isolated from both diseased and control idNs were injected into Xenopus oocytes and both GABA and AMPA currents were successfully recorded. Patch-clamp measurements on idNs revealed depolarized action potential for SCN1AM145T, suggesting a reduced excitability. Expression analyses of VGSCs and chloride co-transporters NKCC1 and KCC2 showed a cellular “dysmaturity” of mutated idNs, strengthened by the high expression of SCN3A, a more fetal-like VGSC isoform, and a high NKCC1/KCC2 ratio, in mutated cells. Overall, we provide strong evidence for an intrinsic cellular immaturity, underscoring the role of mutant NaV1.1 in the development of FS. Furthermore, our data are strengthening previous findings obtained using transfected cells and recordings on human slices, demonstrating that diseased idNs represent a powerful tool for personalized therapy and ex vivo drug screening for human epileptic disorders.
Collapse
Affiliation(s)
- Stefania Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Valeria Lucchino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Luana Scaramuzzino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Pierangelo Cifelli
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of Aquila, 67100 Aquila, Italy;
| | - Tiziano D’Andrea
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
| | | | - Sergio Fucile
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
- IRCCS Neuromed, Via Atinense, 86077 Pozzilli, Italy;
| | - Eleonora Palma
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
| | - Antonio Gambardella
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.G.); (E.I.P.)
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
- IRCCS San Raffaele Roma, Via della Pisana, 00163 Rome, Italy
- Correspondence: (G.R.); (G.C.)
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
- Correspondence: (G.R.); (G.C.)
| | - Elvira Immacolata Parrotta
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.G.); (E.I.P.)
| |
Collapse
|
23
|
Chancey JH, Howard MA. Synaptic Integration in CA1 Pyramidal Neurons Is Intact despite Deficits in GABAergic Transmission in the Scn1a Haploinsufficiency Mouse Model of Dravet Syndrome. eNeuro 2022; 9:ENEURO.0080-22.2022. [PMID: 35523580 PMCID: PMC9116933 DOI: 10.1523/eneuro.0080-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/29/2022] [Accepted: 04/27/2022] [Indexed: 01/13/2023] Open
Abstract
Mutations of SCN1A, which encodes the voltage-gated sodium channel Nav1.1, can cause epilepsy disorders such as Dravet syndrome (DS) that are comorbid with wide-ranging neurologic dysfunction. Many studies suggest that Nav1.1 haploinsufficiency causes forebrain GABAergic interneuron hypoexcitability, while pyramidal neuron physiology is mostly unaltered, and that this serves as a primary cell physiology phenotype linking mutation to disease. We hypothesized that deficits in inhibition would alter synaptic integration during activation of the hippocampal microcircuit, thus disrupting cellular information processing and leading to seizures and cognitive deficits. We tested this hypothesis using ex vivo whole-cell recordings from CA1 pyramidal neurons in a heterozygous Scn1a knock-out mouse model and wild-type (WT) littermates, measuring responses to single and patterned synaptic stimulation and spontaneous synaptic activity. Overall, our experiments reveal a surprising normalcy of excitatory and inhibitory synaptic temporal integration in the hippocampus of Scn1a haploinsufficient mice. While miniature IPSCs and feedforward inhibition and were decreased, we did not identify a pattern or frequency of input that caused a failure of synaptic inhibition. We further show that reduced GABA release probability and subsequent reduced short-term depression may act to overcome deficits in inhibition normalizing input/output functions in the Scn1a haploinsufficient hippocampus. These experiments show that CA1 pyramidal neuron synaptic processing is surprisingly robust, even during decreased interneuron function, and more complex circuit activity is likely required to reveal altered function in the hippocampal microcircuit.
Collapse
Affiliation(s)
- Jessica Hotard Chancey
- Department of Neurology, Dell Medical School, Austin 78712, TX
- Department of Neuroscience and Center for Learning and Memory, University of Texas at Austin, Austin 78712, TX
| | - MacKenzie Allen Howard
- Department of Neurology, Dell Medical School, Austin 78712, TX
- Department of Neuroscience and Center for Learning and Memory, University of Texas at Austin, Austin 78712, TX
| |
Collapse
|
24
|
Interneuronal dynamics facilitate the initiation of spike block in cortical microcircuits. J Comput Neurosci 2022; 50:275-298. [PMID: 35441302 DOI: 10.1007/s10827-022-00815-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 02/09/2022] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Pyramidal cell spike block is a common occurrence in migraine with aura and epileptic seizures. In both cases, pyramidal cells experience hyperexcitation with rapidly increasing firing rates, major changes in electrochemistry, and ultimately spike block that temporarily terminates neuronal activity. In cortical spreading depression (CSD), spike block propagates as a slowly traveling wave of inactivity through cortical pyramidal cells, which is thought to precede migraine attacks with aura. In seizures, highly synchronized cortical activity can be interspersed with, or terminated by, spike block. While the identifying characteristic of CSD and seizures is the pyramidal cell hyperexcitation, it is currently unknown how the dynamics of the cortical microcircuits and inhibitory interneurons affect the initiation of hyperexcitation and subsequent spike block.We tested the contribution of cortical inhibitory interneurons to the initiation of spike block using a cortical microcircuit model that takes into account changes in ion concentrations that result from neuronal firing. Our results show that interneuronal inhibition provides a wider dynamic range to the circuit and generally improves stability against spike block. Despite these beneficial effects, strong interneuronal firing contributed to rapidly changing extracellular ion concentrations, which facilitated hyperexcitation and led to spike block first in the interneuron and then in the pyramidal cell. In all cases, a loss of interneuronal firing triggered pyramidal cell spike block. However, preventing interneuronal spike block was insufficient to rescue the pyramidal cell from spike block. Our data thus demonstrate that while the role of interneurons in cortical microcircuits is complex, they are critical to the initiation of pyramidal cell spike block. We discuss the implications that localized effects on cortical interneurons have beyond the isolated microcircuit and their contribution to CSD and epileptic seizures.
Collapse
|
25
|
Almog Y, Mavashov A, Brusel M, Rubinstein M. Functional Investigation of a Neuronal Microcircuit in the CA1 Area of the Hippocampus Reveals Synaptic Dysfunction in Dravet Syndrome Mice. Front Mol Neurosci 2022; 15:823640. [PMID: 35370551 PMCID: PMC8966673 DOI: 10.3389/fnmol.2022.823640] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/21/2022] [Indexed: 02/05/2023] Open
Abstract
Dravet syndrome is severe childhood-onset epilepsy, caused by loss of function mutations in the SCN1A gene, encoding for the voltage-gated sodium channel NaV1.1. The leading hypothesis is that Dravet is caused by selective reduction in the excitability of inhibitory neurons, due to hampered activity of NaV1.1 channels in these cells. However, these initial neuronal changes can lead to further network alterations. Here, focusing on the CA1 microcircuit in hippocampal brain slices of Dravet syndrome (DS, Scn1aA1783V/WT) and wild-type (WT) mice, we examined the functional response to the application of Hm1a, a specific NaV1.1 activator, in CA1 stratum-oriens (SO) interneurons and CA1 pyramidal excitatory neurons. DS SO interneurons demonstrated reduced firing and depolarized threshold for action potential (AP), indicating impaired activity. Nevertheless, Hm1a induced a similar AP threshold hyperpolarization in WT and DS interneurons. Conversely, a smaller effect of Hm1a was observed in CA1 pyramidal neurons of DS mice. In these excitatory cells, Hm1a application resulted in WT-specific AP threshold hyperpolarization and increased firing probability, with no effect on DS neurons. Additionally, when the firing of SO interneurons was triggered by CA3 stimulation and relayed via activation of CA1 excitatory neurons, the firing probability was similar in WT and DS interneurons, also featuring a comparable increase in the firing probability following Hm1a application. Interestingly, a similar functional response to Hm1a was observed in a second DS mouse model, harboring the nonsense Scn1aR613X mutation. Furthermore, we show homeostatic synaptic alterations in both CA1 pyramidal neurons and SO interneurons, consistent with reduced excitation and inhibition onto CA1 pyramidal neurons and increased release probability in the CA1-SO synapse. Together, these results suggest global neuronal alterations within the CA1 microcircuit extending beyond the direct impact of NaV1.1 dysfunction.
Collapse
Affiliation(s)
- Yael Almog
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Mavashov
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Marina Brusel
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moran Rubinstein
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Moran Rubinstein,
| |
Collapse
|
26
|
Mattis J, Somarowthu A, Goff KM, Jiang E, Yom J, Sotuyo N, Mcgarry LM, Feng H, Kaneko K, Goldberg EM. Corticohippocampal circuit dysfunction in a mouse model of Dravet syndrome. eLife 2022; 11:e69293. [PMID: 35212623 PMCID: PMC8920506 DOI: 10.7554/elife.69293] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Dravet syndrome (DS) is a neurodevelopmental disorder due to pathogenic variants in SCN1A encoding the Nav1.1 sodium channel subunit, characterized by treatment-resistant epilepsy, temperature-sensitive seizures, developmental delay/intellectual disability with features of autism spectrum disorder, and increased risk of sudden death. Convergent data suggest hippocampal dentate gyrus (DG) pathology in DS (Scn1a+/-) mice. We performed two-photon calcium imaging in brain slice to uncover a profound dysfunction of filtering of perforant path input by DG in young adult Scn1a+/- mice. This was not due to dysfunction of DG parvalbumin inhibitory interneurons (PV-INs), which were only mildly impaired at this timepoint; however, we identified enhanced excitatory input to granule cells, suggesting that circuit dysfunction is due to excessive excitation rather than impaired inhibition. We confirmed that both optogenetic stimulation of entorhinal cortex and selective chemogenetic inhibition of DG PV-INs lowered seizure threshold in vivo in young adult Scn1a+/- mice. Optogenetic activation of PV-INs, on the other hand, normalized evoked responses in granule cells in vitro. These results establish the corticohippocampal circuit as a key locus of pathology in Scn1a+/- mice and suggest that PV-INs retain powerful inhibitory function and may be harnessed as a potential therapeutic approach toward seizure modulation.
Collapse
Affiliation(s)
- Joanna Mattis
- Department of Neurology, The Perelman School of Medicine at The University of PennsylvaniaPhiladelphiaUnited States
| | - Ala Somarowthu
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Kevin M Goff
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Evan Jiang
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Jina Yom
- College of Arts and Sciences, The University of PennsylvaniaPhiladelphiaUnited States
| | - Nathaniel Sotuyo
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Laura M Mcgarry
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Huijie Feng
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Keisuke Kaneko
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Ethan M Goldberg
- Department of Neurology, The Perelman School of Medicine at The University of PennsylvaniaPhiladelphiaUnited States
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Neuroscience, The Perelman School of Medicine at The University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
27
|
Godoy LD, Prizon T, Rossignoli MT, Leite JP, Liberato JL. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Front Integr Neurosci 2022; 16:765324. [PMID: 35250498 PMCID: PMC8891758 DOI: 10.3389/fnint.2022.765324] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Parvalbumin is a calcium-binding protein present in inhibitory interneurons that play an essential role in regulating many physiological processes, such as intracellular signaling and synaptic transmission. Changes in parvalbumin expression are deeply related to epilepsy, which is considered one of the most disabling neuropathologies. Epilepsy is a complex multi-factor group of disorders characterized by periods of hypersynchronous activity and hyperexcitability within brain networks. In this scenario, inhibitory neurotransmission dysfunction in modulating excitatory transmission related to the loss of subsets of parvalbumin-expressing inhibitory interneuron may have a prominent role in disrupted excitability. Some studies also reported that parvalbumin-positive interneurons altered function might contribute to psychiatric comorbidities associated with epilepsy, such as depression, anxiety, and psychosis. Understanding the epileptogenic process and comorbidities associated with epilepsy have significantly advanced through preclinical and clinical investigation. In this review, evidence from parvalbumin altered function in epilepsy and associated psychiatric comorbidities were explored with a translational perspective. Some advances in potential therapeutic interventions are highlighted, from current antiepileptic and neuroprotective drugs to cutting edge modulation of parvalbumin subpopulations using optogenetics, designer receptors exclusively activated by designer drugs (DREADD) techniques, transcranial magnetic stimulation, genome engineering, and cell grafting. Creating new perspectives on mechanisms and therapeutic strategies is valuable for understanding the pathophysiology of epilepsy and its psychiatric comorbidities and improving efficiency in clinical intervention.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- João Pereira Leite,
| | - José Luiz Liberato
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: José Luiz Liberato,
| |
Collapse
|
28
|
Studtmann C, Ladislav M, Topolski MA, Safari M, Swanger SA. NaV1.1 haploinsufficiency impairs glutamatergic and GABAergic neuron function in the thalamus. Neurobiol Dis 2022; 167:105672. [DOI: 10.1016/j.nbd.2022.105672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
|
29
|
Beretta S, Gritti L, Ponzoni L, Scalmani P, Mantegazza M, Sala M, Verpelli C, Sala C. Rescuing epileptic and behavioral alterations in a Dravet syndrome mouse model by inhibiting eukaryotic elongation factor 2 kinase (eEF2K). Mol Autism 2022; 13:1. [PMID: 34980259 PMCID: PMC8722032 DOI: 10.1186/s13229-021-00484-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022] Open
Abstract
Background Dravet Syndrome is a severe childhood pharmaco-resistant epileptic disorder mainly caused by mutations in the SCN1A gene, which encodes for the α1 subunit of the type I voltage-gated sodium channel (NaV1.1), that causes imbalance between excitation and inhibition in the brain. We recently found that eEF2K knock out mice displayed enhanced GABAergic transmission and tonic inhibition and were less susceptible to epileptic seizures. Thus, we investigated the effect of inhibition of eEF2K on the epileptic and behavioral phenotype of Scn1a ± mice, a murine model of Dravet Syndrome. Methods To elucidate the role of eEF2K pathway in the etiopathology of Dravet syndrome we generated a new mouse model deleting the eEF2K gene in Scn1a ± mice. By crossing Scn1a ± mice with eEF2K−/− mice we obtained the three main genotypes needed for our studies, Scn1a+/+ eEF2K+/+ (WT mice), Scn1a ± eEF2K+/+ mice (Scn1a ± mice) and Scn1a ± eEF2K−/− mice, that were fully characterized for EEG and behavioral phenotype. Furthermore, we tested the ability of a pharmacological inhibitor of eEF2K in rescuing EEG alterations of the Scn1a ± mice. Results We showed that the activity of eEF2K/eEF2 pathway was enhanced in Scn1a ± mice. Then, we demonstrated that both genetic deletion and pharmacological inhibition of eEF2K were sufficient to ameliorate the epileptic phenotype of Scn1a ± mice. Interestingly we also found that motor coordination defect, memory impairments, and stereotyped behavior of the Scn1a ± mice were reverted by eEF2K deletion. The analysis of spontaneous inhibitory postsynaptic currents (sIPSCs) suggested that the rescue of the pathological phenotype was driven by the potentiation of GABAergic synapses. Limitations Even if we found that eEF2K deletion was able to increase inhibitory synapses function, the molecular mechanism underlining the inhibition of eEF2K/eEF2 pathway in rescuing epileptic and behavioral alterations in the Scn1a ± needs further investigations. Conclusions Our data indicate that pharmacological inhibition of eEF2K could represent a novel therapeutic intervention for treating epilepsy and related comorbidities in the Dravet syndrome. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00484-0.
Collapse
Affiliation(s)
- Stefania Beretta
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Laura Gritti
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Luisa Ponzoni
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Paolo Scalmani
- L'Unità Operativa Complessa di Epilettologia Clinica e Sperimentale, Foundation Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Neurological Institute Carlo Besta, 20133, Milan, Italy
| | - Massimo Mantegazza
- CNRS UMR 7275, Institut National de La Santé Et de La Recherche Médicale, LabEx ICST, Institute of Molecular and Cellular Pharmacology (IPMC), Université Côte d'Azur (UCA), 06560, Valbonne-Sophia Antipolis, France
| | - Mariaelvina Sala
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| | - Carlo Sala
- CNR Neuroscience Institute, Milan, and NeuroMi Milan Center for Neuroscience, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| |
Collapse
|
30
|
Xu C, Zhang Y, Gozal D, Carney P. Channelopathy of Dravet Syndrome and Potential Neuroprotective Effects of Cannabidiol. J Cent Nerv Syst Dis 2021; 13:11795735211048045. [PMID: 34992485 PMCID: PMC8724990 DOI: 10.1177/11795735211048045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dravet syndrome (DS) is a channelopathy, neurodevelopmental, epileptic encephalopathy characterized by seizures, developmental delay, and cognitive impairment that includes susceptibility to thermally induced seizures, spontaneous seizures, ataxia, circadian rhythm and sleep disorders, autistic-like behaviors, and premature death. More than 80% of DS cases are linked to mutations in genes which encode voltage-gated sodium channel subunits, SCN1A and SCN1B, which encode the Nav1.1α subunit and Nav1.1β1 subunit, respectively. There are other gene mutations encoding potassium, calcium, and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels related to DS. One-third of patients have pharmacoresistance epilepsy. DS is unresponsive to standard therapy. Cannabidiol (CBD), a non-psychoactive phytocannabinoid present in Cannabis, has been introduced for treating DS because of its anticonvulsant properties in animal models and humans, especially in pharmacoresistant patients. However, the etiological channelopathiological mechanism of DS and action mechanism of CBD on the channels are unclear. In this review, we summarize evidence of the direct and indirect action mechanism of sodium, potassium, calcium, and HCN channels in DS, especially sodium subunits. Some channels' loss-of-function or gain-of-function in inhibitory or excitatory neurons determine the balance of excitatory and inhibitory are associated with DS. A great variety of mechanisms of CBD anticonvulsant effects are focused on modulating these channels, especially sodium, calcium, and potassium channels, which will shed light on ionic channelopathy of DS and the precise molecular treatment of DS in the future.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Child Health and the Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics; Department of Neuroscience, Uniformed Services University School of Medicine, Bethesda, MD, USA
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Paul Carney
- Departments of Child Health and Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
31
|
Eren-Koçak E, Dalkara T. Ion Channel Dysfunction and Neuroinflammation in Migraine and Depression. Front Pharmacol 2021; 12:777607. [PMID: 34858192 PMCID: PMC8631474 DOI: 10.3389/fphar.2021.777607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/15/2023] Open
Abstract
Migraine and major depression are debilitating disorders with high lifetime prevalence rates. Interestingly these disorders are highly comorbid and show significant heritability, suggesting shared pathophysiological mechanisms. Non-homeostatic function of ion channels and neuroinflammation may be common mechanisms underlying both disorders: The excitation-inhibition balance of microcircuits and their modulation by monoaminergic systems, which depend on the expression and function of membrane located K+, Na+, and Ca+2 channels, have been reported to be disturbed in both depression and migraine. Ion channels and energy supply to synapses not only change excitability of neurons but can also mediate the induction and maintenance of inflammatory signaling implicated in the pathophysiology of both disorders. In this respect, Pannexin-1 and P2X7 large-pore ion channel receptors can induce inflammasome formation that triggers release of pro-inflammatory mediators from the cell. Here, the role of ion channels involved in the regulation of excitation-inhibition balance, synaptic energy homeostasis as well as inflammatory signaling in migraine and depression will be reviewed.
Collapse
Affiliation(s)
- Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Psychiatry, Medical Faculty, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
32
|
Layer N, Sonnenberg L, Pardo González E, Benda J, Hedrich UBS, Lerche H, Koch H, Wuttke TV. Dravet Variant SCN1A A1783V Impairs Interneuron Firing Predominantly by Altered Channel Activation. Front Cell Neurosci 2021; 15:754530. [PMID: 34776868 PMCID: PMC8581729 DOI: 10.3389/fncel.2021.754530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022] Open
Abstract
Dravet syndrome (DS) is a developmental epileptic encephalopathy mainly caused by functional NaV1.1 haploinsufficiency in inhibitory interneurons. Recently, a new conditional mouse model expressing the recurrent human p.(Ala1783Val) missense variant has become available. In this study, we provided an electrophysiological characterization of this variant in tsA201 cells, revealing both altered voltage-dependence of activation and slow inactivation without reduced sodium peak current density. Based on these data, simulated interneuron (IN) firing properties in a conductance-based single-compartment model suggested surprisingly similar firing deficits for NaV1.1A1783V and full haploinsufficiency as caused by heterozygous truncation variants. Impaired NaV1.1A1783V channel activation was predicted to have a significantly larger impact on channel function than altered slow inactivation and is therefore proposed as the main mechanism underlying IN dysfunction. The computational model was validated in cortical organotypic slice cultures derived from conditional Scn1aA1783V mice. Pan-neuronal activation of the p.Ala1783V in vitro confirmed a predicted IN firing deficit and revealed an accompanying reduction of interneuronal input resistance while demonstrating normal excitability of pyramidal neurons. Altered input resistance was fed back into the model for further refinement. Taken together these data demonstrate that primary loss of function (LOF) gating properties accompanied by altered membrane characteristics may match effects of full haploinsufficiency on the neuronal level despite maintaining physiological peak current density, thereby causing DS.
Collapse
Affiliation(s)
- Nikolas Layer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Sonnenberg
- Institute for Neurobiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Emilio Pardo González
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jan Benda
- Institute for Neurobiology, Eberhard Karls University Tübingen, Tübingen, Germany.,Bernstein Center for Computational Neuroscience, Eberhard Karls Universitat, Tübingen, Germany
| | - Ulrike B S Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Henner Koch
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Epileptology, Neurology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| |
Collapse
|
33
|
Auffenberg E, Hedrich UB, Barbieri R, Miely D, Groschup B, Wuttke TV, Vogel N, Lührs P, Zanardi I, Bertelli S, Spielmann N, Gailus-Durner V, Fuchs H, Hrabě de Angelis M, Pusch M, Dichgans M, Lerche H, Gavazzo P, Plesnila N, Freilinger T. Hyperexcitable interneurons trigger cortical spreading depression in an Scn1a migraine model. J Clin Invest 2021; 131:142202. [PMID: 34546973 PMCID: PMC8553559 DOI: 10.1172/jci142202] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cortical spreading depression (CSD), a wave of depolarization followed by depression of cortical activity, is a pathophysiological process implicated in migraine with aura and various other brain pathologies, such as ischemic stroke and traumatic brain injury. To gain insight into the pathophysiology of CSD, we generated a mouse model for a severe monogenic subtype of migraine with aura, familial hemiplegic migraine type 3 (FHM3). FHM3 is caused by mutations in SCN1A, encoding the voltage-gated Na+ channel NaV1.1 predominantly expressed in inhibitory interneurons. Homozygous Scn1aL1649Q knock-in mice died prematurely, whereas heterozygous mice had a normal lifespan. Heterozygous Scn1aL1649Q knock-in mice compared with WT mice displayed a significantly enhanced susceptibility to CSD. We found L1649Q to cause a gain-of-function effect with an impaired Na+-channel inactivation and increased ramp Na+ currents leading to hyperactivity of fast-spiking inhibitory interneurons. Brain slice recordings using K+-sensitive electrodes revealed an increase in extracellular K+ in the early phase of CSD in heterozygous mice, likely representing the mechanistic link between interneuron hyperactivity and CSD initiation. The neuronal phenotype and premature death of homozygous Scn1aL1649Q knock-in mice was partially rescued by GS967, a blocker of persistent Na+ currents. Collectively, our findings identify interneuron hyperactivity as a mechanism to trigger CSD.
Collapse
Affiliation(s)
- Eva Auffenberg
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Ulrike Bs Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Raffaella Barbieri
- Biophysics Institute, Consiglio Nazionale delle Ricerche (CNR), Genoa, Italy
| | - Daniela Miely
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Bernhard Groschup
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Niklas Vogel
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Philipp Lührs
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ilaria Zanardi
- Biophysics Institute, Consiglio Nazionale delle Ricerche (CNR), Genoa, Italy
| | - Sara Bertelli
- Biophysics Institute, Consiglio Nazionale delle Ricerche (CNR), Genoa, Italy
| | - Nadine Spielmann
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Michael Pusch
- Biophysics Institute, Consiglio Nazionale delle Ricerche (CNR), Genoa, Italy
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Paola Gavazzo
- Biophysics Institute, Consiglio Nazionale delle Ricerche (CNR), Genoa, Italy
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tobias Freilinger
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Department of Neurology, Klinikum Passau, Passau, Germany
| |
Collapse
|
34
|
Chever O, Zerimech S, Scalmani P, Lemaire L, Pizzamiglio L, Loucif A, Ayrault M, Krupa M, Desroches M, Duprat F, Léna I, Cestèle S, Mantegazza M. Initiation of migraine-related cortical spreading depolarization by hyperactivity of GABAergic neurons and NaV1.1 channels. J Clin Invest 2021; 131:e142203. [PMID: 34491914 PMCID: PMC8553565 DOI: 10.1172/jci142203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/02/2021] [Indexed: 01/24/2023] Open
Abstract
Spreading depolarizations (SDs) are involved in migraine, epilepsy, stroke, traumatic brain injury, and subarachnoid hemorrhage. However, the cellular origin and specific differential mechanisms are not clear. Increased glutamatergic activity is thought to be the key factor for generating cortical spreading depression (CSD), a pathological mechanism of migraine. Here, we show that acute pharmacological activation of NaV1.1 (the main Na+ channel of interneurons) or optogenetic-induced hyperactivity of GABAergic interneurons is sufficient to ignite CSD in the neocortex by spiking-generated extracellular K+ build-up. Neither GABAergic nor glutamatergic synaptic transmission were required for CSD initiation. CSD was not generated in other brain areas, suggesting that this is a neocortex-specific mechanism of CSD initiation. Gain-of-function mutations of NaV1.1 (SCN1A) cause familial hemiplegic migraine type-3 (FHM3), a subtype of migraine with aura, of which CSD is the neurophysiological correlate. Our results provide the mechanism linking NaV1.1 gain of function to CSD generation in FHM3. Thus, we reveal the key role of hyperactivity of GABAergic interneurons in a mechanism of CSD initiation, which is relevant as a pathological mechanism of Nav1.1 FHM3 mutations, and possibly also for other types of migraine and diseases in which SDs are involved.
Collapse
Affiliation(s)
- Oana Chever
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Sarah Zerimech
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Paolo Scalmani
- Unità Operativa VII Clinical and Experimental Epileptology, Foundation IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | - Louisiane Lemaire
- Inria Sophia Antipolis Méditerranée, MathNeuro Project Team, Valbonne-Sophia Antipolis, France
| | - Lara Pizzamiglio
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Alexandre Loucif
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Marion Ayrault
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Martin Krupa
- Université Côte d'Azur, Laboratoire Jean-Alexandre Dieudonné, Nice, France
| | - Mathieu Desroches
- Inria Sophia Antipolis Méditerranée, MathNeuro Project Team, Valbonne-Sophia Antipolis, France
| | - Fabrice Duprat
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| | - Isabelle Léna
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Massimo Mantegazza
- Université Côte d'Azur and.,CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| |
Collapse
|
35
|
Bagheri S, Haddadi R, Saki S, Kourosh-Arami M, Komaki A. The effect of sodium channels on neurological/neuronal disorders: A systematic review. Int J Dev Neurosci 2021; 81:669-685. [PMID: 34687079 DOI: 10.1002/jdn.10153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/06/2021] [Accepted: 10/19/2021] [Indexed: 12/19/2022] Open
Abstract
Neurological and neuronal disorders are associated with structural, biochemical, or electrical abnormalities in the nervous system. Many neurological diseases have not yet been discovered. Interventions used for the treatment of these disorders include avoidance measures, lifestyle changes, physiotherapy, neurorehabilitation, pain management, medication, and surgery. In the sodium channelopathies, alterations in the structure, expression, and function of voltage-gated sodium channels (VGSCs) are considered as the causes of neurological and neuronal diseases. Online databases, including Scopus, Science Direct, Google Scholar, and PubMed were assessed for studies published between 1977 and 2020 using the keywords of review, sodium channels blocker, neurological diseases, and neuronal diseases. VGSCs consist of one α subunit and two β subunits. These subunits are known to regulate the gating kinetics, functional characteristics, and localization of the ion channel. These channels are involved in cell migration, cellular connections, neuronal pathfinding, and neurite outgrowth. Through the VGSC, the action potential is triggered and propagated in the neurons. Action potentials are physiological functions and passage of impermeable ions. The electrophysiological properties of these channels and their relationship with neurological and neuronal disorders have been identified. Subunit mutations are involved in the development of diseases, such as epilepsy, multiple sclerosis, autism, and Alzheimer's disease. Accordingly, we conducted a review of the link between VGSCs and neurological and neuronal diseases. Also, novel therapeutic targets were introduced for future drug discoveries.
Collapse
Affiliation(s)
- Shokufeh Bagheri
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology, School of Pharmacy, Hamadan University of Medical Science, Hamadan, Iran
| | - Sahar Saki
- Vice-Chancellor for Research and Technology, Hamadan University of Medical Science, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
36
|
Hedrich UBS, Lauxmann S, Wolff M, Synofzik M, Bast T, Binelli A, Serratosa JM, Martínez-Ulloa P, Allen NM, King MD, Gorman KM, Zeev BB, Tzadok M, Wong-Kisiel L, Marjanovic D, Rubboli G, Sisodiya SM, Lutz F, Ashraf HP, Torge K, Yan P, Bosselmann C, Schwarz N, Fudali M, Lerche H. 4-Aminopyridine is a promising treatment option for patients with gain-of-function KCNA2-encephalopathy. Sci Transl Med 2021; 13:eaaz4957. [PMID: 34516822 DOI: 10.1126/scitranslmed.aaz4957] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ulrike B S Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Stephan Lauxmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Markus Wolff
- Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital, 72076 Tuebingen, Germany.,Department of Pediatric Neurology, Vivantes-Klinikum Neukölln, 12351 Berlin, Germany
| | - Matthis Synofzik
- Department of Neurology and Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Thomas Bast
- Epilepsy Center Kork, 77694 Kehl-Kork, Germany.,Medical Faculty of the University of Freiburg, 79110 Freiburg, Germany
| | - Adrian Binelli
- Department of Pediatric Neurology, Elizalde Children's Hospital, C1270 Buenos Aires, Argentina
| | - José M Serratosa
- Neurology Laboratory and Epilepsy Unit, Department of Neurology, IIS- Fundacio'n Jime'nez Dı'az, UAM, 28040 Madrid, Spain.,Centro de Investigacio'n Biome'dica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Pedro Martínez-Ulloa
- Neurology Laboratory and Epilepsy Unit, Department of Neurology, IIS- Fundacio'n Jime'nez Dı'az, UAM, 28040 Madrid, Spain
| | - Nicholas M Allen
- Department of Paediatrics, Clinical Sciences Institute, National University of Ireland Galway, Galway H91 TK33, Ireland
| | - Mary D King
- Department of Neurology and Neurophysiology, Children's Health Ireland at Temple Street, Dublin DO1 YC67, Ireland.,School of Medicine and Medical Science, University College Dublin, Dublin DO4 V1W8, Ireland
| | - Kathleen M Gorman
- Department of Neurology and Neurophysiology, Children's Health Ireland at Temple Street, Dublin DO1 YC67, Ireland.,School of Medicine and Medical Science, University College Dublin, Dublin DO4 V1W8, Ireland
| | - Bruria Ben Zeev
- Sackler School of Medicine Tel Aviv University, Tel Aviv 6997801, Israel.,Pediatric Neurology Unit, Edmond and Lilly Safra Pediatric Hospital, Sheba Medical Center, 5265601 Ramat Gan, Israel
| | - Michal Tzadok
- Sackler School of Medicine Tel Aviv University, Tel Aviv 6997801, Israel.,Pediatric Neurology Unit, Edmond and Lilly Safra Pediatric Hospital, Sheba Medical Center, 5265601 Ramat Gan, Israel
| | - Lily Wong-Kisiel
- Divisions of Child Neurology & Division of Epilepsy, Department of Neurology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | - Guido Rubboli
- Danish Epilepsy Center, Filadelfia, 4293 Dianalund, Denmark.,University of Copenhagen, 1165 Copenhagen, Denmark
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.,Chalfont Centre for Epilepsy, Bucks SL9 0RJ, UK
| | - Florian Lutz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Harshad Pannikkaveettil Ashraf
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Kirsten Torge
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Pu Yan
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Christian Bosselmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Monika Fudali
- Department of Neurosurgery, University of Tuebingen, 72076 Tuebingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
37
|
Lemaire L, Desroches M, Krupa M, Pizzamiglio L, Scalmani P, Mantegazza M. Modeling NaV1.1/SCN1A sodium channel mutations in a microcircuit with realistic ion concentration dynamics suggests differential GABAergic mechanisms leading to hyperexcitability in epilepsy and hemiplegic migraine. PLoS Comput Biol 2021; 17:e1009239. [PMID: 34314446 PMCID: PMC8345895 DOI: 10.1371/journal.pcbi.1009239] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 08/06/2021] [Accepted: 07/02/2021] [Indexed: 11/19/2022] Open
Abstract
Loss of function mutations of SCN1A, the gene coding for the voltage-gated sodium channel NaV1.1, cause different types of epilepsy, whereas gain of function mutations cause sporadic and familial hemiplegic migraine type 3 (FHM-3). However, it is not clear yet how these opposite effects can induce paroxysmal pathological activities involving neuronal networks’ hyperexcitability that are specific of epilepsy (seizures) or migraine (cortical spreading depolarization, CSD). To better understand differential mechanisms leading to the initiation of these pathological activities, we used a two-neuron conductance-based model of interconnected GABAergic and pyramidal glutamatergic neurons, in which we incorporated ionic concentration dynamics in both neurons. We modeled FHM-3 mutations by increasing the persistent sodium current in the interneuron and epileptogenic mutations by decreasing the sodium conductance in the interneuron. Therefore, we studied both FHM-3 and epileptogenic mutations within the same framework, modifying only two parameters. In our model, the key effect of gain of function FHM-3 mutations is ion fluxes modification at each action potential (in particular the larger activation of voltage-gated potassium channels induced by the NaV1.1 gain of function), and the resulting CSD-triggering extracellular potassium accumulation, which is not caused only by modifications of firing frequency. Loss of function epileptogenic mutations, on the other hand, increase GABAergic neurons’ susceptibility to depolarization block, without major modifications of firing frequency before it. Our modeling results connect qualitatively to experimental data: potassium accumulation in the case of FHM-3 mutations and facilitated depolarization block of the GABAergic neuron in the case of epileptogenic mutations. Both these effects can lead to pyramidal neuron hyperexcitability, inducing in the migraine condition depolarization block of both the GABAergic and the pyramidal neuron. Overall, our findings suggest different mechanisms of network hyperexcitability for migraine and epileptogenic NaV1.1 mutations, implying that the modifications of firing frequency may not be the only relevant pathological mechanism. The voltage-gated sodium channel NaV1.1 is a major target of human mutations implicated in different pathologies. In particular, mutations identified in certain types of epilepsy cause loss of function of the channel, whereas mutations identified in certain types of migraine (in which spreading depolarizations of the cortical circuits of the brain are involved) cause instead gain of function. Here, we study dysfunctions induced by these differential effects in a two-neuron (GABAergic and pyramidal) conductance-based model with dynamic ion concentrations. We obtain results that can be related to experimental findings in both situations. Namely, extracellular potassium accumulation induced by the activity of the GABAergic neuron in the case of CSD, and higher propensity of the GABAergic neuron to depolarization block in the epileptogenic scenario, without significant modifications of its firing frequency prior to it. Both scenarios can induce hyperexcitability of the pyramidal neuron, leading in the migraine condition to depolarization block of both the GABAergic and the pyramidal neuron. Our results are successfully confronted to experimental data and suggest that modification of firing frequency is not the only key mechanism in these pathologies of neuronal excitability.
Collapse
Affiliation(s)
- Louisiane Lemaire
- Inria Sophia Antipolis Méditerranée Research Centre, MathNeuro Team, Valbonne-Sophia Antipolis, France
- Université Côte d’Azur, Nice, France
- * E-mail: (LL); (MM)
| | - Mathieu Desroches
- Inria Sophia Antipolis Méditerranée Research Centre, MathNeuro Team, Valbonne-Sophia Antipolis, France
- Université Côte d’Azur, Nice, France
| | - Martin Krupa
- Inria Sophia Antipolis Méditerranée Research Centre, MathNeuro Team, Valbonne-Sophia Antipolis, France
- Université Côte d’Azur, Laboratoire Jean-Alexandre Dieudonné, Nice, France
| | - Lara Pizzamiglio
- Université Côte d’Azur, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
- CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
| | - Paolo Scalmani
- U.O. VII Clinical and Experimental Epileptology, Foundation IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | - Massimo Mantegazza
- Université Côte d’Azur, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
- CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France
- Inserm, Valbonne-Sophia Antipolis, France
- * E-mail: (LL); (MM)
| |
Collapse
|
38
|
Binini N, Talpo F, Spaiardi P, Maniezzi C, Pedrazzoli M, Raffin F, Mattiello N, Castagno AN, Masetto S, Yanagawa Y, Dickson CT, Ramat S, Toselli M, Biella GR. Membrane Resonance in Pyramidal and GABAergic Neurons of the Mouse Perirhinal Cortex. Front Cell Neurosci 2021; 15:703407. [PMID: 34366789 PMCID: PMC8339929 DOI: 10.3389/fncel.2021.703407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
The perirhinal cortex (PRC) is a polymodal associative region of the temporal lobe that works as a gateway between cortical areas and hippocampus. In recent years, an increasing interest arose in the role played by the PRC in learning and memory processes, such as object recognition memory, in contrast with certain forms of hippocampus-dependent spatial and episodic memory. The integrative properties of the PRC should provide all necessary resources to select and enhance the information to be propagated to and from the hippocampus. Among these properties, we explore in this paper the ability of the PRC neurons to amplify the output voltage to current input at selected frequencies, known as membrane resonance. Within cerebral circuits the resonance of a neuron operates as a filter toward inputs signals at certain frequencies to coordinate network activity in the brain by affecting the rate of neuronal firing and the precision of spike timing. Furthermore, the ability of the PRC neurons to resonate could have a fundamental role in generating subthreshold oscillations and in the selection of cortical inputs directed to the hippocampus. Here, performing whole-cell patch-clamp recordings from perirhinal pyramidal neurons and GABAergic interneurons of GAD67-GFP+ mice, we found, for the first time, that the majority of PRC neurons are resonant at their resting potential, with a resonance frequency of 0.5–1.5 Hz at 23°C and of 1.5–2.8 Hz at 36°C. In the presence of ZD7288 (blocker of HCN channels) resonance was abolished in both pyramidal neurons and interneurons, suggesting that Ih current is critically involved in resonance generation. Otherwise, application of TTx (voltage-dependent Na+ channel blocker) attenuates the resonance in pyramidal neurons but not in interneurons, suggesting that only in pyramidal neurons the persistent sodium current has an amplifying effect. These experimental results have also been confirmed by a computational model. From a functional point of view, the resonance in the PRC would affect the reverberating activity between neocortex and hippocampus, especially during slow wave sleep, and could be involved in the redistribution and strengthening of memory representation in cortical regions.
Collapse
Affiliation(s)
- Noemi Binini
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Paolo Spaiardi
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Claudia Maniezzi
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Matteo Pedrazzoli
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Francesca Raffin
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Niccolò Mattiello
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Antonio N Castagno
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Sergio Masetto
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University, Maebashi, Japan
| | - Clayton T Dickson
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| | - Stefano Ramat
- Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Mauro Toselli
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Gerardo Rosario Biella
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| |
Collapse
|
39
|
Ahnaou A, Drinkenburg WHIM. Sleep, neuronal hyperexcitability, inflammation and neurodegeneration: Does early chronic short sleep trigger and is it the key to overcoming Alzheimer's disease? Neurosci Biobehav Rev 2021; 129:157-179. [PMID: 34214513 DOI: 10.1016/j.neubiorev.2021.06.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 05/13/2021] [Accepted: 06/25/2021] [Indexed: 01/13/2023]
Abstract
Evidence links neuroinflammation to Alzheimer's disease (AD); however, its exact contribution to the onset and progression of the disease is poorly understood. Symptoms of AD can be seen as the tip of an iceberg, consisting of a neuropathological build-up in the brain of extracellular amyloid-β (Aβ) plaques and intraneuronal hyperphosphorylated aggregates of Tau (pTau), which are thought to stem from an imbalance between its production and clearance resulting in loss of synaptic health and dysfunctional cortical connectivity. The glymphatic drainage system, which is particularly active during sleep, plays a key role in the clearance of proteinopathies. Poor sleep can cause hyperexcitability and promote Aβ and tau pathology leading to systemic inflammation. The early neuronal hyperexcitability of γ-aminobutyric acid (GABA)-ergic inhibitory interneurons and impaired inhibitory control of cortical pyramidal neurons lie at the crossroads of excitatory/inhibitory imbalance and inflammation. We outline, with a prospective framework, a possible vicious spiral linking early chronic short sleep, neuronal hyperexcitability, inflammation and neurodegeneration. Understanding the early predictors of AD, through an integrative approach, may hold promise for reducing attrition in the late stages of neuroprotective drug development.
Collapse
Affiliation(s)
- A Ahnaou
- Dept. of Neuroscience Discovery, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, B-2340, Belgium.
| | - W H I M Drinkenburg
- Dept. of Neuroscience Discovery, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, B-2340, Belgium
| |
Collapse
|
40
|
Uchino K, Kawano H, Tanaka Y, Adaniya Y, Asahara A, Deshimaru M, Kubota K, Watanabe T, Katsurabayashi S, Iwasaki K, Hirose S. Inhibitory synaptic transmission is impaired at higher extracellular Ca 2+ concentrations in Scn1a +/- mouse model of Dravet syndrome. Sci Rep 2021; 11:10634. [PMID: 34017040 PMCID: PMC8137694 DOI: 10.1038/s41598-021-90224-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Dravet syndrome (DS) is an intractable form of childhood epilepsy that occurs in infancy. More than 80% of all patients have a heterozygous abnormality in the SCN1A gene, which encodes a subunit of Na+ channels in the brain. However, the detailed pathogenesis of DS remains unclear. This study investigated the synaptic pathogenesis of this disease in terms of excitatory/inhibitory balance using a mouse model of DS. We show that excitatory postsynaptic currents were similar between Scn1a knock-in neurons (Scn1a+/- neurons) and wild-type neurons, but inhibitory postsynaptic currents were significantly lower in Scn1a+/- neurons. Moreover, both the vesicular release probability and the number of inhibitory synapses were significantly lower in Scn1a+/- neurons compared with wild-type neurons. There was no proportional increase in inhibitory postsynaptic current amplitude in response to increased extracellular Ca2+ concentrations. Our study revealed that the number of inhibitory synapses is significantly reduced in Scn1a+/- neurons, while the sensitivity of inhibitory synapses to extracellular Ca2+ concentrations is markedly increased. These data suggest that Ca2+ tethering in inhibitory nerve terminals may be disturbed following the synaptic burst, likely leading to epileptic symptoms.
Collapse
Affiliation(s)
- Kouya Uchino
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Hiroyuki Kawano
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Yasuyoshi Tanaka
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Yuna Adaniya
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Ai Asahara
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Masanobu Deshimaru
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan.
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shinichi Hirose
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
- Department of Pediatrics, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
41
|
Izsak J, Seth H, Iljin M, Theiss S, Ågren H, Funa K, Aigner L, Hanse E, Illes S. Differential acute impact of therapeutically effective and overdose concentrations of lithium on human neuronal single cell and network function. Transl Psychiatry 2021; 11:281. [PMID: 33980815 PMCID: PMC8115174 DOI: 10.1038/s41398-021-01399-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023] Open
Abstract
Lithium salts are used as mood-balancing medication prescribed to patients suffering from neuropsychiatric disorders, such as bipolar disorder and major depressive disorder. Lithium salts cross the blood-brain barrier and reach the brain parenchyma within few hours after oral application, however, how lithium influences directly human neuronal function is unknown. We applied patch-clamp and microelectrode array technology on human induced pluripotent stem cell (iPSC)-derived cortical neurons acutely exposed to therapeutic (<1 mM) and overdose concentrations (>1 mM) of lithium chloride (LiCl) to assess how therapeutically effective and overdose concentrations of LiCl directly influence human neuronal electrophysiological function at the synapse, single-cell, and neuronal network level. We describe that human iPSC-cortical neurons exposed to lithium showed an increased neuronal activity under all tested concentrations. Furthermore, we reveal a lithium-induced, concentration-dependent, transition of regular synchronous neuronal network activity using therapeutically effective concentration (<1 mM LiCl) to epileptiform-like neuronal discharges using overdose concentration (>1 mM LiCl). The overdose concentration lithium-induced epileptiform-like activity was similar to the epileptiform-like activity caused by the GABAA-receptor antagonist. Patch-clamp recordings reveal that lithium reduces action potential threshold at all concentrations, however, only overdose concentration causes increased frequency of spontaneous AMPA-receptor mediated transmission. By applying the AMPA-receptor antagonist and anti-epileptic drug Perampanel, we demonstrate that Perampanel suppresses lithium-induced epileptiform-like activity in human cortical neurons. We provide insights in how therapeutically effective and overdose concentration of lithium directly influences human neuronal function at synapse, a single neuron, and neuronal network levels. Furthermore, we provide evidence that Perampanel suppresses pathological neuronal discharges caused by overdose concentrations of lithium in human neurons.
Collapse
Affiliation(s)
- Julia Izsak
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Henrik Seth
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Margarita Iljin
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Stephan Theiss
- grid.411327.20000 0001 2176 9917Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany ,Result Medical GmbH, Düsseldorf, Germany
| | - Hans Ågren
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Keiko Funa
- grid.8761.80000 0000 9919 9582Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XOncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ludwig Aigner
- grid.21604.310000 0004 0523 5263Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Eric Hanse
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Illes
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
42
|
Izsak J, Seth H, Theiss S, Hanse E, Illes S. Human Cerebrospinal Fluid Promotes Neuronal Circuit Maturation of Human Induced Pluripotent Stem Cell-Derived 3D Neural Aggregates. Stem Cell Reports 2021; 14:1044-1059. [PMID: 32521247 PMCID: PMC7355159 DOI: 10.1016/j.stemcr.2020.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 01/09/2023] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived in vitro neural and organoid models resemble fetal, rather than adult brain properties, indicating that currently applied cultivation media and supplements are insufficient to achieve neural maturation beyond the fetal stage. In vivo, cerebrospinal fluid molecules are regulating the transition of the immature fetal human brain into a mature adult brain. By culturing hiPSC-3D neural aggregates in human cerebrospinal fluid (hCSF) obtained from healthy adult individuals, we demonstrate that hCSF rapidly triggers neurogenesis, gliogenesis, synapse formation, neurite outgrowth, suppresses proliferation of residing neural stem cells, and results in the formation of synchronously active neuronal circuits in vitro within 3 days. Thus, a physiologically relevant and adult brain-like milieu triggers maturation of hiPSC-3D neural aggregates into highly functional neuronal circuits in vitro. The approach presented here opens a new avenue to identify novel physiological factors for the improvement of hiPSC neural in vitro models. Human CSF triggers rapidly multiple maturation processes in human 3D neural models Human CSF triggers human neurogenesis and suppresses neural stem cell proliferation Human CSF triggers human astrocyte development, neurite growth, and synapse formation Human CSF triggers the maturation of neurons into highly functional neuronal circuits
Collapse
Affiliation(s)
- Julia Izsak
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Henrik Seth
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Stephan Theiss
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany; Result Medical GmbH, Düsseldorf, Germany
| | - Eric Hanse
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Illes
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
43
|
Epilepsy and Migraine Shared Genetic and Molecular Mechanisms: Focus on Therapeutic Strategies. Mol Neurobiol 2021; 58:3874-3883. [PMID: 33856647 DOI: 10.1007/s12035-021-02386-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Epilepsy and migraine are both episodic disorders and share clinical as well as pathophysiological mechanisms. The prevalence of epilepsy in migraine patients is generally higher than normal as compared to general population and vice versa. Various environmental risk factors and genetic factors have been reported to be associated with susceptibility of these comorbid diseases. Specific genes have been implicated in the pathogenesis of the two diseases. However, the shared genetic susceptibility has not been explored extensively. Previous studies have reported that the alterations in the genes encoding ion channel proteins are common risk factors for both the diseases. The alterations in ion channel-encoding genes CACNAIA (T666M) and SCNIA (Q1489K and L1649Q) have been found to be involved in the development of familial hemiplegic migraine (FHM) as well as generalized epilepsy and some cases of focal epilepsy as well. The fact that both these disorders are treated with anti-epileptic drugs (AEDs) strongly supports common underlying mechanisms. This review has been compiled with an aim to explore the alterations in common genes involved in various pathways regulating neuronal hyperexcitability, a common risk factor for both these conditions. The avenue for future treatment strategies targeting common genes and molecular mechanisms has also been discussed.
Collapse
|
44
|
Chen W, Luo B, Gao N, Li H, Wang H, Li L, Cui W, Zhang L, Sun D, Liu F, Dong Z, Ren X, Zhang H, Su H, Xiong WC, Mei L. Neddylation stabilizes Nav1.1 to maintain interneuron excitability and prevent seizures in murine epilepsy models. J Clin Invest 2021; 131:136956. [PMID: 33651714 DOI: 10.1172/jci136956] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
The excitability of interneurons requires Nav1.1, the α subunit of the voltage-gated sodium channel. Nav1.1 deficiency and mutations reduce interneuron excitability, a major pathological mechanism for epilepsy syndromes. However, the regulatory mechanisms of Nav1.1 expression remain unclear. Here, we provide evidence that neddylation is critical to Nav1.1 stability. Mutant mice lacking Nae1, an obligatory component of the E1 ligase for neddylation, in parvalbumin-positive interneurons (PVINs) exhibited spontaneous epileptic seizures and premature death. Electrophysiological studies indicate that Nae1 deletion reduced PVIN excitability and GABA release and consequently increased the network excitability of pyramidal neurons (PyNs). Further analysis revealed a reduction in sodium-current density, not a change in channel property, in mutant PVINs and decreased Nav1.1 protein levels. These results suggest that insufficient neddylation in PVINs reduces Nav1.1 stability and thus the excitability of PVINs; the ensuing increased PyN activity causes seizures in mice. Consistently, Nav1.1 was found reduced by proteomic analysis that revealed abnormality in synapses and metabolic pathways. Our findings describe a role of neddylation in maintaining Nav1.1 stability for PVIN excitability and reveal what we believe is a new mechanism in the pathogenesis of epilepsy.
Collapse
Affiliation(s)
- Wenbing Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nannan Gao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Haiwen Li
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Lei Li
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wanpeng Cui
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Lei Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fang Liu
- Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Huabo Su
- Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Interneuron Dysfunction in a New Mouse Model of SCN1A GEFS. eNeuro 2021; 8:ENEURO.0394-20.2021. [PMID: 33658306 PMCID: PMC8174035 DOI: 10.1523/eneuro.0394-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 11/21/2022] Open
Abstract
Advances in genome sequencing have identified over 1300 mutations in the SCN1A sodium channel gene that result in genetic epilepsies. However, it still remains unclear how most individual mutations within SCN1A result in seizures. A previous study has shown that the K1270T (KT) mutation, linked to genetic epilepsy with febrile seizure plus (GEFS+) in humans, causes heat-induced seizure activity associated with a temperature-dependent decrease in GABAergic neuron excitability in a Drosophila knock-in model. To examine the behavioral and cellular effects of this mutation in mammals, we introduced the equivalent KT mutation into the mouse (Mus musculus) Scn1a (Scn1aKT) gene using CRISPR/Cas9 and generated mutant lines in two widely used genetic backgrounds: C57BL/6NJ and 129X1/SvJ. In both backgrounds, mice homozygous for the KT mutation had spontaneous seizures and died by postnatal day (P)23. There was no difference in mortality of heterozygous KT mice compared with wild-type littermates up to six months old. Heterozygous mutants exhibited heat-induced seizures at ∼42°C, a temperature that did not induce seizures in wild-type littermates. In acute hippocampal slices at permissive temperatures, current-clamp recordings revealed a significantly depolarized shift in action potential threshold and reduced action potential amplitude in parvalbumin (PV)-expressing inhibitory CA1 interneurons in Scn1aKT/+ mice. There was no change in the firing properties of excitatory CA1 pyramidal neurons. These results suggest that a constitutive decrease in inhibitory interneuron excitability contributes to the seizure phenotype in the mouse model.
Collapse
|
46
|
Mantegazza M, Cestèle S, Catterall WA. Sodium channelopathies of skeletal muscle and brain. Physiol Rev 2021; 101:1633-1689. [PMID: 33769100 DOI: 10.1152/physrev.00025.2020] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels initiate action potentials in nerve, skeletal muscle, and other electrically excitable cells. Mutations in them cause a wide range of diseases. These channelopathy mutations affect every aspect of sodium channel function, including voltage sensing, voltage-dependent activation, ion conductance, fast and slow inactivation, and both biosynthesis and assembly. Mutations that cause different forms of periodic paralysis in skeletal muscle were discovered first and have provided a template for understanding structure, function, and pathophysiology at the molecular level. More recent work has revealed multiple sodium channelopathies in the brain. Here we review the well-characterized genetics and pathophysiology of the periodic paralyses of skeletal muscle and then use this information as a foundation for advancing our understanding of mutations in the structurally homologous α-subunits of brain sodium channels that cause epilepsy, migraine, autism, and related comorbidities. We include studies based on molecular and structural biology, cell biology and physiology, pharmacology, and mouse genetics. Our review reveals unexpected connections among these different types of sodium channelopathies.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France
| | | |
Collapse
|
47
|
Altered Fast Synaptic Transmission in a Mouse Model of DNM1-Associated Developmental Epileptic Encephalopathy. eNeuro 2021; 8:ENEURO.0269-20.2020. [PMID: 33372033 PMCID: PMC7986544 DOI: 10.1523/eneuro.0269-20.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/05/2022] Open
Abstract
Developmental epileptic encephalopathies (DEEs) are severe seizure disorders that occur in infants and young children, characterized by developmental delay, cognitive decline, and early mortality. Recent efforts have identified a wide variety of genetic variants that cause DEEs. Among these, variants in the DNM1 gene have emerged as definitive causes of DEEs, including infantile spasms and Lennox–Gastaut syndrome. A mouse model of Dnm1-associated DEE, known as “Fitful” (Dnm1Ftfl), recapitulates key features of the disease, including spontaneous seizures, early lethality, and neuronal degeneration. Previous work showed that DNM1 is a key regulator of synaptic vesicle (SV) endocytosis and synaptic transmission and suggested that inhibitory neurotransmission may be more reliant on DNM1 function than excitatory transmission. The Dnm1Ftfl variant is thought to encode a dominant negative DNM1 protein; however, the effects of the Dnm1Ftfl variant on synaptic transmission are largely unknown. To understand these synaptic effects, we recorded from pairs of cultured mouse cortical neurons and characterized all four major connection types [excitation of excitation (E-E), inhibition of inhibition (I-I), E-I, I-E]. Miniature and spontaneous EPSCs and IPSCs were larger, but less frequent, at all Dnm1Ftfl synaptic types, and Dnm1Ftfl neurons had reduced expression of excitatory and inhibitory SV markers. Baseline evoked transmission, however, was reduced only at inhibitory synapses onto excitatory neurons, because of a smaller pool of releasable SVs. In addition to these synaptic alterations, Dnm1Ftfl neurons degenerated later in development, although their activity levels were reduced, suggesting that Dnm1Ftfl may impair synaptic transmission and neuronal health through distinct mechanisms.
Collapse
|
48
|
Voskobiynyk Y, Battu G, Felker SA, Cochran JN, Newton MP, Lambert LJ, Kesterson RA, Myers RM, Cooper GM, Roberson ED, Barsh GS. Aberrant regulation of a poison exon caused by a non-coding variant in a mouse model of Scn1a-associated epileptic encephalopathy. PLoS Genet 2021; 17:e1009195. [PMID: 33411788 PMCID: PMC7790302 DOI: 10.1371/journal.pgen.1009195] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/14/2020] [Indexed: 12/20/2022] Open
Abstract
Dravet syndrome (DS) is a developmental and epileptic encephalopathy that results from mutations in the Nav1.1 sodium channel encoded by SCN1A. Most known DS-causing mutations are in coding regions of SCN1A, but we recently identified several disease-associated SCN1A mutations in intron 20 that are within or near to a cryptic and evolutionarily conserved "poison" exon, 20N, whose inclusion is predicted to lead to transcript degradation. However, it is not clear how these intron 20 variants alter SCN1A expression or DS pathophysiology in an organismal context, nor is it clear how exon 20N is regulated in a tissue-specific and developmental context. We address those questions here by generating an animal model of our index case, NM_006920.4(SCN1A):c.3969+2451G>C, using gene editing to create the orthologous mutation in laboratory mice. Scn1a heterozygous knock-in (+/KI) mice exhibited an ~50% reduction in brain Scn1a mRNA and Nav1.1 protein levels, together with characteristics observed in other DS mouse models, including premature mortality, seizures, and hyperactivity. In brain tissue from adult Scn1a +/+ animals, quantitative RT-PCR assays indicated that ~1% of Scn1a mRNA included exon 20N, while brain tissue from Scn1a +/KI mice exhibited an ~5-fold increase in the extent of exon 20N inclusion. We investigated the extent of exon 20N inclusion in brain during normal fetal development in RNA-seq data and discovered that levels of inclusion were ~70% at E14.5, declining progressively to ~10% postnatally. A similar pattern exists for the homologous sodium channel Nav1.6, encoded by Scn8a. For both genes, there is an inverse relationship between the level of functional transcript and the extent of poison exon inclusion. Taken together, our findings suggest that poison exon usage by Scn1a and Scn8a is a strategy to regulate channel expression during normal brain development, and that mutations recapitulating a fetal-like pattern of splicing cause reduced channel expression and epileptic encephalopathy.
Collapse
Affiliation(s)
- Yuliya Voskobiynyk
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer’s Disease Center, and Evelyn F. McKnight Brain Institute, Departments, of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Gopal Battu
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
| | - Stephanie A. Felker
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
- Department of Department of Biotechnology Science and Engineering, University of Alabama in Huntsville, Hunstville, AL, United States AL, United States of America
| | - J. Nicholas Cochran
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
| | - Megan P. Newton
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Laura J. Lambert
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Robert A. Kesterson
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Richard M. Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
| | - Gregory M. Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
| | - Erik D. Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer’s Disease Center, and Evelyn F. McKnight Brain Institute, Departments, of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail: (GSB); (EDR)
| | - Gregory S. Barsh
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
- * E-mail: (GSB); (EDR)
| |
Collapse
|
49
|
Vezyroglou A, Varadkar S, Bast T, Hirsch E, Strobl K, Harvey AS, Scheffer IE, Sisodiya SM, Cross JH. Focal epilepsy in SCN1A-mutation carrying patients: is there a role for epilepsy surgery? Dev Med Child Neurol 2020; 62:1331-1335. [PMID: 32538476 DOI: 10.1111/dmcn.14588] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2020] [Indexed: 11/28/2022]
Abstract
Variants in the gene SCN1A are a common genetic cause for a wide range of epilepsy phenotypes ranging from febrile seizures to Dravet syndrome. Focal onset seizures and structural lesions can be present in these patients and the question arises whether epilepsy surgery should be considered. We report eight patients (mean age 13y 11mo [SD 8y 1mo], range 3-26y; four females, four males) with SCN1A variants, who underwent epilepsy surgery. Outcomes were variable and seemed to be directly related to the patient's anatomo-electroclinical epilepsy phenotype. Patients with Dravet syndrome had unfavourable outcomes, whilst patients with focal epilepsy, proven to arise from a single structural lesion, had good results. We conclude that the value of epilepsy surgery in patients with an SCN1A variant rests on two issues: understanding whether the variant is pathogenic and the patient's anatomo-electroclinical phenotype. Careful evaluation of epilepsy phenotype integrated with understanding the significance of genetic variants is essential in determining a patient's suitability for epilepsy surgery. Patients with focal onset epilepsy may benefit from epilepsy surgery, whereas those with Dravet syndrome do not. WHAT THIS PAPER ADDS: Patients should not automatically be excluded from epilepsy surgery evaluation if they carry an SCN1A variant. Patients with focal epilepsy may benefit from epilepsy surgery; those with Dravet syndrome do not.
Collapse
Affiliation(s)
- Aikaterini Vezyroglou
- Department of Developmental Neurosciences, UCL NIHR BRC Great Ormond Street Institute of Child Health, London, UK.,Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Sophia Varadkar
- Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Thomas Bast
- Kork Epilepsy Center, Kehl-Kork, Germany.,Medical Faculty of the University of Freiburg, Freiburg, Germany
| | - Edouard Hirsch
- Medical and Surgical Epilepsy Unit, Hautepierre Hospital, University of Strasbourg, Strasbourg, France
| | | | - A Simon Harvey
- Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | | | - Ingrid E Scheffer
- Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia.,Florey Institute and Murdoch Children's Research Institute, Austin Health and Royal Children's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - J Helen Cross
- Department of Developmental Neurosciences, UCL NIHR BRC Great Ormond Street Institute of Child Health, London, UK.,Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
50
|
Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T, Ishii A. Application of induced pluripotent stem cells in epilepsy. Mol Cell Neurosci 2020; 108:103535. [DOI: 10.1016/j.mcn.2020.103535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
|