1
|
Pizzamiglio L, Capitano F, Rusina E, Fossati G, Menna E, Léna I, Antonucci F, Mantegazza M. Neurodevelopmental defects in Dravet syndrome Scn1a +/- mice: Targeting GABA-switch rescues behavioral dysfunctions but not seizures and mortality. Neurobiol Dis 2025; 207:106853. [PMID: 40021096 DOI: 10.1016/j.nbd.2025.106853] [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: 12/26/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Dravet syndrome (DS) is a developmental and epileptic encephalopathy (DEE) caused by mutations of the SCN1A gene (NaV1.1 sodium channel) and characterized by seizures, motor disabilities and cognitive/behavioral deficits, including autistic traits. The relative role of seizures and neurodevelopmental defects in disease progression, as well as the role of the mutation in inducing early neurodevelopmental defects before symptoms' onset, are not clear yet. A delayed switch of GABAergic transmission from excitatory to inhibitory (GABA-switch) was reported in models of DS, but its effects on the phenotype have not been investigated. Using a multi-scale approach, here we show that targeting GABA-switch with the drugs KU55933 (KU) or bumetanide (which upregulate KCC2 or inhibits NKCC1 chloride transporters, respectively) rescues social interaction deficits and reduces hyperactivity observed in P21 Scn1a+/- DS mouse model. Bumetanide also improves spatial working memory defects. Importantly, neither KU nor bumetanide have effect on seizures or mortality rate. Also, we disclose early behavioral defects and delayed neurodevelopmental milestones well before seizure onset, at the beginning of NaV1.1 expression. We thus reveal that neurodevelopmental components in DS, in particular GABA switch, underlie some cognitive/behavioral defects, but not seizures. Our work provides further evidence that seizures and neuropsychiatric dysfunctions in DEEs can be uncoupled and can have differential pathological mechanisms. They could be treated separately with targeted pharmacological strategies.
Collapse
Affiliation(s)
- Lara Pizzamiglio
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | - Fabrizio Capitano
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | - Evgeniia Rusina
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | | | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Rozzano, Milan, Italy
| | - Isabelle Léna
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France
| | - Flavia Antonucci
- Institute of Neuroscience - National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Rozzano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy.
| | - Massimo Mantegazza
- Université Côte d'Azur, Valbonne-Sophia Antipolis, France; CNRS UMR7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne-Sophia Antipolis, France; Inserm U1323, Valbonne-Sophia Antipolis, France.
| |
Collapse
|
2
|
Mich JK, Ryu J, Wei AD, Gore BB, Guo R, Bard AM, Martinez RA, Luber EM, Liu J, Bishaw YM, Christian RJ, Oliveira LM, Miranda N, Ramirez JM, Ting JT, Lein ES, Levi BP, Kalume FK. Interneuron-specific dual-AAV SCN1A gene replacement corrects epileptic phenotypes in mouse models of Dravet syndrome. Sci Transl Med 2025; 17:eadn5603. [PMID: 40106582 DOI: 10.1126/scitranslmed.adn5603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
Dravet syndrome (DS) is a severe developmental epileptic encephalopathy marked by treatment-resistant seizures, developmental delay, intellectual disability, motor deficits, and a 10 to 20% rate of premature death. Most patients with DS harbor loss-of-function mutations in one copy of SCN1A, which encodes the voltage-gated sodium channel (NaV)1.1 alpha subunit and has been associated with inhibitory neuron dysfunction. Here, we generated a split-intein form of SCN1A and used a dual-vector delivery approach to circumvent adeno-associated virus (AAV) packaging limitations. In addition, we applied previously developed enhancer technology to produce an interneuron-specific gene replacement therapy for DS, called DLX2.0-SCN1A. The split-intein SCN1A vectors produced full-length NaV1.1 protein, and functional sodium channels were recorded in HEK293 cells in vitro. Administration of dual DLX2.0-SCN1A AAVs to wild-type mice produced full-length, reconstituted human protein by Western blot and telencephalic interneuron-specific and dose-dependent NaV1.1 expression by immunohistochemistry. These vectors also conferred strong dose-dependent protection against postnatal mortality and seizures in Scn1afl/+;Meox2-Cre and Scn1a+/R613X DS mouse models. Injection of single or dual DLX2.0-SCN1A AAVs into wild-type mice did not result in increased mortality, weight loss, or gliosis as measured by immunohistochemistry. In contrast, expression of SCN1A in all neurons driven by the human SYNAPSIN I promoter caused an adverse effect marked by increased mortality in the preweaning period, before disease onset. These findings demonstrate proof of concept that interneuron-specific AAV-mediated SCN1A gene replacement can rescue DS phenotypes in mouse models and suggest that it could be a therapeutic approach for patients with DS.
Collapse
Affiliation(s)
- John K Mich
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jiyun Ryu
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Aguan D Wei
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Bryan B Gore
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rong Guo
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Angela M Bard
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Emily M Luber
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jiatai Liu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Luiz M Oliveira
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Jan-Marino Ramirez
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Franck K Kalume
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98104, USA
| |
Collapse
|
3
|
Lee SH, Kang YJ, Smith BN. Activation of hypoactive parvalbumin-positive fast-spiking interneurons restores dentate inhibition to reduce electrographic seizures in the mouse intrahippocampal kainate model of temporal lobe epilepsy. Neurobiol Dis 2024; 203:106737. [PMID: 39542222 DOI: 10.1016/j.nbd.2024.106737] [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: 04/05/2024] [Revised: 10/20/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
Parvalbumin-positive (PV+) GABAergic interneurons in the dentate gyrus provide powerful perisomatic inhibition of dentate granule cells (DGCs) to prevent overexcitation and maintain the stability of dentate gyrus circuits. Most dentate PV+ interneurons survive status epilepticus, but surviving PV+ interneuron mediated inhibition is compromised in the dentate gyrus shortly after status epilepticus, contributing to epileptogenesis in temporal lobe epilepsy. It is uncertain whether the impaired activity of dentate PV+ interneurons recovers at later times or if it continues for months following status epilepticus. The development of compensatory modifications related to PV+ interneuron circuits in the months following status epilepticus is unknown, although reduced dentate GABAergic inhibition persists long after status epilepticus. We employed whole-cell patch-clamp recordings from dentate PV+ interneurons and DGCs in slices from male and female sham controls and intrahippocampal kainate (IHK) treated mice that developed spontaneous seizures months after status epilepticus to study epilepsy-associated changes in dentate PV+ interneuron circuits. Electrical recordings showed that: 1) Action potential firing rates of dentate PV+ interneurons were reduced in IHK treated mice up to four months after status epilepticus; 2) spontaneous inhibitory postsynaptic currents (sIPSCs) in DGCs exhibited reduced frequency but increased amplitude in IHK treated mice; and 3) the amplitude of IPSCs in DGCs evoked by optogenetic activation of dentate PV+ cells was upregulated without changes in short-term plasticity. Video-EEG recordings revealed that IHK treated mice showed spontaneous electrographic seizures in the dentate gyrus and that chemogenetic activation of PV+ interneurons abolished electrographic seizures. Our results suggest not only that the compensatory changes in PV+ interneuron circuits develop after IHK treatment, but also that increased PV+ interneuron mediated inhibition in the dentate gyrus may compensate for cell loss and reduced intrinsic excitability of dentate PV+ interneurons to stop seizures in temporal lobe epilepsy.
Collapse
Affiliation(s)
- Sang-Hun Lee
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Young-Jin Kang
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Bret N Smith
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
4
|
Zhu L, Demetriou Y, Barden J, Disla J, Mattis J. Medial septum parvalbumin-expressing inhibitory neurons are impaired in a mouse model of Dravet Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620933. [PMID: 39554146 PMCID: PMC11565850 DOI: 10.1101/2024.10.29.620933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Dravet syndrome (DS) is a severe neurodevelopmental disorder caused by pathogenic variants in the SCN1A gene, which encodes the voltage-gated sodium channel Na v 1.1 α subunit. Experiments in animal models of DS - including the haploinsufficient Scn1a +/- mouse - have identified impaired excitability of interneurons in the hippocampus and neocortex; this is thought to underlie the treatment-resistant epilepsy that is a prominent feature of the DS phenotype. However, additional brain structures, such as the medial septum (MS), also express SCN1A . The medial septum is known to play an important role in cognitive function and thus may contribute to the intellectual impairment that also characterizes DS. In this study, we employed whole cell patch clamp recordings in acute brain slices to characterize the electrophysiological properties of MS neurons in Scn1a +/- mice versus age-matched wild-type littermate controls. We found no discernible genotype-related differences in MS cholinergic (ChAT) neurons, but significant dysfunction within MS parvalbumin-expressing (PV) inhibitory neurons in Scn1a +/- mice. We further identified heterogeneity of firing patterns among MS PV neurons, and additional genotype differences in the proportion of subtype representation. These results confirm that the MS is an additional locus of pathology in DS, that may contribute to co- morbidities such as cognitive impairment.
Collapse
|
5
|
Miralles RM, Boscia AR, Kittur S, Hanflink JC, Panchal PS, Yorek MS, Deutsch TCJ, Reever CM, Vundela SR, Wengert ER, Patel MK. Parvalbumin interneuron impairment causes synaptic transmission deficits and seizures in SCN8A developmental and epileptic encephalopathy. JCI Insight 2024; 9:e181005. [PMID: 39435659 PMCID: PMC11529981 DOI: 10.1172/jci.insight.181005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/05/2024] [Indexed: 10/23/2024] Open
Abstract
SCN8A developmental and epileptic encephalopathy (DEE) is a severe epilepsy syndrome resulting from mutations in the voltage-gated sodium channel Nav1.6, encoded by the gene SCN8A. Nav1.6 is expressed in excitatory and inhibitory neurons, yet previous studies primarily focus on how SCN8A mutations affect excitatory neurons, with limited studies on the importance of inhibitory interneurons. Parvalbumin (PV) interneurons are a prominent inhibitory interneuron subtype that expresses Nav1.6. To assess PV interneuron function within SCN8A DEE, we used 2 mouse models harboring patient-derived SCN8A gain-of-function variants, Scn8aD/+, where the SCN8A variant N1768D is expressed globally, and Scn8aW/+-PV, where the SCN8A variant R1872W is selectively expressed in PV interneurons. Expression of the R1872W SCN8A variant selectively in PV interneurons led to development of spontaneous seizures and seizure-induced death. Electrophysiology studies showed that Scn8aD/+ and Scn8aW/+-PV interneurons were susceptible to depolarization block and exhibited increased persistent sodium current. Evaluation of synaptic connections between PV interneurons and pyramidal cells showed synaptic transmission deficits in Scn8aD/+ and Scn8aW/+-PV interneurons. Together, our findings indicate that PV interneuron failure via depolarization block along with inhibitory synaptic impairment likely elicits an overall inhibitory reduction in SCN8A DEE, leading to unchecked excitation and ultimately resulting in seizures and seizure-induced death.
Collapse
Affiliation(s)
- Raquel M. Miralles
- Department of Anesthesiology and
- Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, Virginia, USA
| | | | | | | | | | | | | | - Caeley M. Reever
- Department of Anesthesiology and
- Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, Virginia, USA
| | | | - Eric R. Wengert
- Department of Anesthesiology and
- Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, Virginia, USA
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Manoj K. Patel
- Department of Anesthesiology and
- Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
6
|
Wengert ER, Cheng MA, Liebergall SR, Markwalter KH, Hong Y, Arias L, Marsh ED, Zhang X, Somarowthu A, Goldberg EM. Impaired excitability of fast-spiking neurons in a novel mouse model of KCNC1 epileptic encephalopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615463. [PMID: 39386579 PMCID: PMC11463657 DOI: 10.1101/2024.09.27.615463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The recurrent pathogenic variant KCNC1-p.Ala421Val (A421V) is a cause of developmental and epileptic encephalopathy characterized by moderate-to-severe developmental delay/intellectual disability, and infantile-onset treatment-resistant epilepsy with multiple seizure types including myoclonic seizures. Yet, the mechanistic basis of disease is unclear. KCNC1 encodes Kv3.1, a voltage-gated potassium channel subunit that is highly and selectively expressed in neurons capable of generating action potentials at high frequency, including parvalbumin-positive fast-spiking GABAergic inhibitory interneurons in cerebral cortex (PV-INs) known to be important for cognitive function and plasticity as well as control of network excitation to prevent seizures. In this study, we generate a novel transgenic mouse model with conditional expression of the Ala421Val pathogenic missense variant (Kcnc1-A421V/+ mice) to explore the physiological mechanisms of KCNC1 developmental and epileptic encephalopathy. Our results indicate that global heterozygous expression of the A421V variant leads to epilepsy and premature lethality. We observe decreased PV-IN cell surface expression of Kv3.1 via immunohistochemistry, decreased voltage-gated potassium current density in PV-INs using outside-out nucleated macropatch recordings in brain slice, and profound impairments in the intrinsic excitability of cerebral cortex PV-INs but not excitatory neurons in current-clamp electrophysiology. In vivo two-photon calcium imaging revealed hypersynchronous discharges correlated with brief paroxysmal movements, subsequently shown to be myoclonic seizures on electroencephalography. We found alterations in PV-IN-mediated inhibitory neurotransmission in young adult but not juvenile Kcnc1-A421V/+ mice relative to wild-type controls. Together, these results establish the impact of the recurrent Kv3.1-A421V variant on neuronal excitability and synaptic physiology across development to drive network dysfunction underlying KCNC1 epileptic encephalopathy.
Collapse
Affiliation(s)
- Eric R. Wengert
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Melody A. Cheng
- School of Arts and Sciences, The University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Sophie R. Liebergall
- The Medical Scientist Training Program, The University of Pennsylvania Perelman School of Medicine, Philadelphia, U.S.A
- Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, U.S.A
| | - Kelly H. Markwalter
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Yerahm Hong
- School of Engineering and Applied Sciences, The University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Leroy Arias
- School of Arts and Sciences, The University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Eric D. Marsh
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, U.S.A
| | - Xiaohong Zhang
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Ala Somarowthu
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Ethan M. Goldberg
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- The Epilepsy Neurogenetics Initiative, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- The Center for Brain Research in Development, Genetics, and Engineering (BRIDGE), The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, U.S.A
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, U.S.A
| |
Collapse
|
7
|
Liebergall SR, Goldberg EM. Ndnf Interneuron Excitability Is Spared in a Mouse Model of Dravet Syndrome. J Neurosci 2024; 44:e1977232024. [PMID: 38443186 PMCID: PMC11044195 DOI: 10.1523/jneurosci.1977-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 03/07/2024] Open
Abstract
Dravet syndrome (DS) is a neurodevelopmental disorder characterized by epilepsy, developmental delay/intellectual disability, and features of autism spectrum disorder, caused by heterozygous loss-of-function variants in SCN1A encoding the voltage-gated sodium channel α subunit Nav1.1. The dominant model of DS pathogenesis is the "interneuron hypothesis," whereby GABAergic interneurons (INs) express and preferentially rely on Nav1.1-containing sodium channels for action potential (AP) generation. This has been shown for three of the major subclasses of cerebral cortex GABAergic INs: those expressing parvalbumin (PV), somatostatin, and vasoactive intestinal peptide. Here, we define the function of a fourth major subclass of INs expressing neuron-derived neurotrophic factor (Ndnf) in male and female DS (Scn1a+/-) mice. Patch-clamp electrophysiological recordings of Ndnf-INs in brain slices from Scn1a+/â mice and WT controls reveal normal intrinsic membrane properties, properties of AP generation and repetitive firing, and synaptic transmission across development. Immunohistochemistry shows that Nav1.1 is strongly expressed at the axon initial segment (AIS) of PV-expressing INs but is absent at the Ndnf-IN AIS. In vivo two-photon calcium imaging demonstrates that Ndnf-INs in Scn1a+/â mice are recruited similarly to WT controls during arousal. These results suggest that Ndnf-INs are the only major IN subclass that does not prominently rely on Nav1.1 for AP generation and thus retain their excitability in DS. The discovery of a major IN subclass with preserved function in the Scn1a+/â mouse model adds further complexity to the "interneuron hypothesis" and highlights the importance of considering cell-type heterogeneity when investigating mechanisms underlying neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sophie R Liebergall
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
- Medical Scientist Training Program, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Ethan M Goldberg
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
- Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
- The Epilepsy Neurogenetics Initiative, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| |
Collapse
|
8
|
Whyte-Fagundes PA, Vance A, Carroll A, Figueroa F, Manukyan C, Baraban SC. Testing of putative antiseizure medications in a preclinical Dravet syndrome zebrafish model. Brain Commun 2024; 6:fcae135. [PMID: 38707709 PMCID: PMC11069116 DOI: 10.1093/braincomms/fcae135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 05/07/2024] Open
Abstract
Dravet syndrome is a severe genetic epilepsy primarily caused by de novo mutations in a voltage-activated sodium channel gene (SCN1A). Patients face life-threatening seizures that are largely resistant to available anti-seizure medications. Preclinical Dravet syndrome animal models are a valuable tool to identify candidate anti-seizure medications for these patients. Among these, scn1lab mutant zebrafish, exhibiting spontaneous seizure-like activity, are particularly amenable to large-scale drug screening. Thus far, we have screened more than 3000 drug candidates in scn1lab zebrafish mutants, identifying valproate, stiripentol, and fenfluramine e.g. Food and Drug Administration-approved drugs, with clinical application in the Dravet syndrome population. Successful phenotypic screening in scn1lab mutant zebrafish is rigorous and consists of two stages: (i) a locomotion-based assay measuring high-velocity convulsive swim behaviour and (ii) an electrophysiology-based assay, using in vivo local field potential recordings, to quantify electrographic seizure-like events. Historically, nearly 90% of drug candidates fail during translation from preclinical models to the clinic. With such a high failure rate, it becomes necessary to address issues of replication and false positive identification. Leveraging our scn1lab zebrafish assays is one approach to address these problems. Here, we curated a list of nine anti-seizure drug candidates recently identified by other groups using preclinical Dravet syndrome models: 1-Ethyl-2-benzimidazolinone, AA43279, chlorzoxazone, donepezil, lisuride, mifepristone, pargyline, soticlestat and vorinostat. First-stage locomotion-based assays in scn1lab mutant zebrafish identified only 1-Ethyl-2-benzimidazolinone, chlorzoxazone and lisuride. However, second-stage local field potential recording assays did not show significant suppression of spontaneous electrographic seizure activity for any of the nine anti-seizure drug candidates. Surprisingly, soticlestat induced frank electrographic seizure-like discharges in wild-type control zebrafish. Taken together, our results failed to replicate clear anti-seizure efficacy for these drug candidates highlighting a necessity for strict scientific standards in preclinical identification of anti-seizure medications.
Collapse
Affiliation(s)
- Paige A Whyte-Fagundes
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Anjelica Vance
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Aloe Carroll
- Behavioral Neurosciences, Northeastern University, Boston, MA 02115, USA
| | - Francisco Figueroa
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Catherine Manukyan
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Scott C Baraban
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
9
|
Lee SH, Kang YJ, Smith BN. Activation of hypoactive parvalbumin-positive fast-spiking interneuron restores dentate inhibition to prevent epileptiform activity in the mouse intrahippocampal kainate model of temporal lobe epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588316. [PMID: 38645248 PMCID: PMC11030452 DOI: 10.1101/2024.04.05.588316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Parvalbumin-positive (PV+) GABAergic interneurons in the dentate gyrus provide powerful perisomatic inhibition of dentate granule cells (DGCs) to prevent overexcitation and maintain the stability of dentate gyrus circuits. Most dentate PV+ interneurons survive status epilepticus, but surviving PV+ interneuron mediated inhibition is compromised in the dentate gyrus shortly after status epilepticus, contributing to epileptogenesis in temporal lobe epilepsy. It is uncertain whether the impaired activity of dentate PV+ interneurons recovers at later times or if it continues for months following status epilepticus. The development of compensatory modifications related to PV+ interneuron circuits in the months following status epilepticus is unknown, although reduced dentate GABAergic inhibition persists long after status epilepticus. We employed PV immunostaining and whole-cell patch-clamp recordings from dentate PV+ interneurons and DGCs in slices from male and female sham controls and intrahippocampal kainate (IHK) treated mice that developed spontaneous seizures months after status epilepticus to study epilepsy-associated changes in dentate PV+ interneuron circuits. We found that the number of dentate PV+ cells was reduced in IHK treated mice. Electrical recordings showed that: 1) Action potential firing rates of dentate PV+ interneurons were reduced in IHK treated mice up to four months after status epilepticus; 2) Spontaneous inhibitory postsynaptic currents (sIPSCs) in DGCs exhibited reduced frequency but increased amplitude in IHK treated mice; and 3) The amplitude of evoked IPSCs in DGCs by optogenetic activation of dentate PV+ cells was upregulated without changes in short-term plasticity. Video-EEG recordings revealed that IHK treated mice showed spontaneous epileptiform activity in the dentate gyrus and that chemogenetic activation of PV+ interneurons abolished the epileptiform activity. Our results suggest not only that the compensatory changes in PV+ interneuron circuits develop after IHK treatment, but also that increased PV+ interneuron mediated inhibition in the dentate gyrus may compensate for cell loss and reduced intrinsic excitability of dentate PV+ interneurons to stop seizures in temporal lobe epilepsy. Highlights Reduced number of dentate PV+ interneurons in TLE micePersistently reduced action potential firing rates of dentate PV+ interneurons in TLE miceEnhanced amplitude but decreased frequency of spontaneous IPSCs in the dentate gyrus in TLE miceIncreased amplitude of evoked IPSCs mediated by dentate PV+ interneurons in TLE miceChemogenetic activation of PV+ interneurons prevents epileptiform activity in TLE mice.
Collapse
|
10
|
Miralles RM, Boscia AR, Kittur S, Vundela SR, Wengert ER, Patel MK. Parvalbumin Interneuron Impairment Leads to Synaptic Transmission Deficits and Seizures in SCN8A Epileptic Encephalopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579511. [PMID: 38464208 PMCID: PMC10925130 DOI: 10.1101/2024.02.09.579511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
SCN8A epileptic encephalopathy (EE) is a severe epilepsy syndrome resulting from de novo mutations in the voltage-gated sodium channel Na v 1.6, encoded by the gene SCN8A . Na v 1.6 is expressed in both excitatory and inhibitory neurons, yet previous studies have primarily focused on the impact SCN8A mutations have on excitatory neuron function, with limited studies on the importance of inhibitory interneurons to seizure onset and progression. Inhibitory interneurons are critical in balancing network excitability and are known to contribute to the pathophysiology of other epilepsies. Parvalbumin (PV) interneurons are the most prominent inhibitory neuron subtype in the brain, making up about 40% of inhibitory interneurons. Notably, PV interneurons express high levels of Na v 1.6. To assess the role of PV interneurons within SCN8A EE, we used two mouse models harboring patient-derived SCN8A gain-of-function mutations, Scn8a D/+ , where the SCN8A mutation N1768D is expressed globally, and Scn8a W/+ -PV, where the SCN8A mutation R1872W is selectively expressed in PV interneurons. Expression of the R1872W SCN8A mutation selectively in PV interneurons led to the development of spontaneous seizures in Scn8a W/+ -PV mice and seizure-induced death, decreasing survival compared to wild-type. Electrophysiology studies showed that PV interneurons in Scn8a D/+ and Scn8a W/+ -PV mice were susceptible to depolarization block, a state of action potential failure. Scn8a D/+ and Scn8a W/+ -PV interneurons also exhibited increased persistent sodium current, a hallmark of SCN8A gain-of-function mutations that contributes to depolarization block. Evaluation of synaptic connections between PV interneurons and pyramidal cells showed an increase in synaptic transmission failure at high frequencies (80-120Hz) as well as an increase in synaptic latency in Scn8a D/+ and Scn8a W/+ -PV interneurons. These data indicate a distinct impairment of synaptic transmission in SCN8A EE, potentially decreasing overall cortical network inhibition. Together, our novel findings indicate that failure of PV interneuron spiking via depolarization block along with frequency-dependent inhibitory synaptic impairment likely elicits an overall reduction in the inhibitory drive in SCN8A EE, leading to unchecked excitation and ultimately resulting in seizures and seizure-induced death.
Collapse
|
11
|
Cha J, Filatov G, Smith SJ, Gammaitoni AR, Lothe A, Reeder T. Fenfluramine increases survival and reduces markers of neurodegeneration in a mouse model of Dravet syndrome. Epilepsia Open 2024; 9:300-313. [PMID: 38018342 PMCID: PMC10839300 DOI: 10.1002/epi4.12873] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE In patients with Dravet syndrome (DS), fenfluramine reduced convulsive seizure frequency and provided clinical benefit in nonseizure endpoints (e.g., executive function, survival). In zebrafish mutant scn1 DS models, chronic fenfluramine treatment preserved neuronal cytoarchitecture prior to seizure onset and prevented gliosis; here, we extend these findings to a mammalian model of DS (Scn1a+/- mice) by evaluating the effects of fenfluramine on neuroinflammation (degenerated myelin, activated microglia) and survival. METHODS Scn1a+/- DS mice were treated subcutaneously once daily with fenfluramine (15 mg/kg) or vehicle from postnatal day (PND) 7 until 35-37. Sagittal brain sections were processed for immunohistochemistry using antibodies to degraded myelin basic protein (D-MBP) for degenerated myelin, or CD11b for activated (inflammatory) microglia; sections were scored semi-quantitatively. Apoptotic nuclei were quantified by TUNEL assay. Statistical significance was evaluated by 1-way ANOVA with post-hoc Dunnett's test (D-MBP, CD11b, and TUNEL) or Logrank Mantel-Cox (survival). RESULTS Quantitation of D-MBP immunostaining per 0.1 mm2 unit area of the parietal cortex and hippocampus CA3 yielded significantly higher spheroidal and punctate myelin debris counts in vehicle-treated DS mice than in wild-type mice. Fenfluramine treatment in DS mice significantly reduced these counts. Activated CD11b + microglia were more abundant in DS mouse corpus callosum and hippocampus than in wild-type controls. Fenfluramine treatment of DS mice resulted in significantly fewer activated CD11b + microglia than vehicle-treated DS mice in these brain regions. TUNEL staining in corpus callosum was increased in DS mice relative to wild-type controls. Fenfluramine treatment in DS mice lowered TUNEL staining relative to vehicle-treated DS mice. By PND 35-37, 55% of control DS mice had died, compared with 24% of DS mice receiving fenfluramine treatment (P = 0.0291). SIGNIFICANCE This is the first report of anti-neuroinflammation and pro-survival after fenfluramine treatment in a mammalian DS model. These results corroborate prior data in humans and animal models and suggest important pharmacological activities for fenfluramine beyond seizure reduction. PLAIN LANGUAGE SUMMARY Dravet syndrome is a severe epilepsy disorder that impairs learning and causes premature death. Clinical studies in patients with Dravet syndrome show that fenfluramine reduces convulsive seizures. Additional studies suggest that fenfluramine may have benefits beyond seizures, including promoting survival and improving control over emotions and behavior. Our study is the first to use a Dravet mouse model to investigate nonseizure outcomes of fenfluramine. Results showed that fenfluramine treatment of Dravet mice reduced neuroinflammation significantly more than saline treatment. Fenfluramine-treated Dravet mice also lived longer than saline-treated mice. These results support clinical observations that fenfluramine may have benefits beyond seizures.
Collapse
Affiliation(s)
- John Cha
- University of California San FranciscoSan FranciscoCaliforniaUSA
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
| | - Gregory Filatov
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
- Crosshair Therapeutics, Inc.SunnyvaleCaliforniaUSA
| | - Steven J. Smith
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
- WuXi AppTec, Inc.San FranciscoCaliforniaUSA
| | | | | | - Thadd Reeder
- Zogenix, Inc. (now a part of UCB)EmeryvilleCaliforniaUSA
| |
Collapse
|
12
|
Miralles R, Patel MK. Unspooling the Thread: VIP Interneurons Linked With Autism Spectrum Disorder Behaviors but Not Seizures in Dravet Syndrome. Epilepsy Curr 2024; 24:62-64. [PMID: 38327541 PMCID: PMC10846521 DOI: 10.1177/15357597231218876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
VIP Interneuron Impairment Promotes In Vivo Circuit Dysfunction and Autism-Related Behaviors in Dravet Syndrome Goff KM, Liebergall SR, Jiang E, Somarowthu A, Goldberg EM. Cell Rep . 2023;42(6):112628. doi:10.1016/j.celrep.2023.112628 Dravet syndrome (DS) is a severe neurodevelopmental disorder caused by loss-of-function variants in SCN1A, which encodes the voltage-gated sodium channel subunit Nav1.1. We recently showed that neocortical vasoactive intestinal peptide interneurons (VIP-INs) express Nav1.1 and are hypoexcitable in DS (Scn1a+/-) mice. Here, we investigate VIP-IN function at the circuit and behavioral level by performing in vivo 2-photon calcium imaging in awake wild-type (WT) and Scn1a+/- mice. VIP-IN and pyramidal neuron activation during behavioral transition from quiet wakefulness to active running is diminished in Scn1a+/- mice, and optogenetic activation of VIP-INs restores pyramidal neuron activity to WT levels during locomotion. VIP-IN selective Scn1a deletion reproduces core autism-spectrum-disorder-related behaviors in addition to cellular- and circuit-level deficits in VIP-IN function, but without epilepsy, sudden death, or avoidance behaviors seen in the global model. Hence, VIP-INs are impaired in vivo, which may underlie non-seizure cognitive and behavioral comorbidities in DS.
Collapse
Affiliation(s)
- Raquel Miralles
- Department of Anesthesiology University of Virginia Health System
| | - Manoj K Patel
- Department of Anesthesiology University of Virginia Health System
| |
Collapse
|
13
|
Teralı K, Türkyılmaz A, Sağer SG, Çebi AH. Prediction of molecular phenotypes for novel SCN1A variants from a Turkish genetic epilepsy syndromes cohort and report of two new patients with recessive Dravet syndrome. Clin Transl Sci 2024; 17:e13679. [PMID: 37955180 PMCID: PMC10772300 DOI: 10.1111/cts.13679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023] Open
Abstract
Dravet syndrome and genetic epilepsy with febrile seizures plus (GEFS+) are both epilepsy syndromes that can be attributed to deleterious mutations occurring in SCN1A, the gene encoding the pore-forming α-subunit of the NaV 1.1 voltage-gated sodium channel predominantly expressed in the central nervous system. In this research endeavor, our goal is to expand our prior cohort of Turkish patients affected by SCN1A-positive genetic epilepsy disorders. This will be accomplished by incorporating two recently discovered and infrequent index cases who possess a novel biallelic (homozygous) SCN1A missense variant, namely E158G, associated with Dravet syndrome. Furthermore, our intention is to use computational techniques to predict the molecular phenotypes of each distinct SCN1A variant that has been detected to date within our center. The correlation between genotype and phenotype in Dravet syndrome/GEFS+ is intricate and necessitates meticulous clinical investigation as well as advanced scientific exploration. Broadened mechanistic and structural insights into NaV 1.1 dysfunction offer significant promise in facilitating the development of targeted and effective therapies, which will ultimately enhance clinical outcomes in the treatment of epilepsy.
Collapse
Affiliation(s)
- Kerem Teralı
- Department of Medical Biochemistry, Faculty of MedicineCyprus International UniversityNicosiaCyprus
| | - Ayberk Türkyılmaz
- Department of Medical Genetics, Faculty of MedicineKaradeniz Technical UniversityTrabzonTurkey
| | - Safiye Güneş Sağer
- Department of Pediatric NeurologyKartal Dr. Lütfi Kırdar City HospitalİstanbulTurkey
| | - Alper Han Çebi
- Department of Medical Genetics, Faculty of MedicineKaradeniz Technical UniversityTrabzonTurkey
| |
Collapse
|
14
|
Mich JK, Ryu J, Wei AD, Gore BB, Guo R, Bard AM, Martinez RA, Bishaw Y, Luber E, Oliveira Santos LM, Miranda N, Ramirez JM, Ting JT, Lein ES, Levi BP, Kalume FK. AAV-mediated interneuron-specific gene replacement for Dravet syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571820. [PMID: 38168178 PMCID: PMC10760176 DOI: 10.1101/2023.12.15.571820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Dravet syndrome (DS) is a devastating developmental epileptic encephalopathy marked by treatment-resistant seizures, developmental delay, intellectual disability, motor deficits, and a 10-20% rate of premature death. Most DS patients harbor loss-of-function mutations in one copy of SCN1A , which has been associated with inhibitory neuron dysfunction. Here we developed an interneuron-targeting AAV human SCN1A gene replacement therapy using cell class-specific enhancers. We generated a split-intein fusion form of SCN1A to circumvent AAV packaging limitations and deliver SCN1A via a dual vector approach using cell class-specific enhancers. These constructs produced full-length Na V 1.1 protein and functional sodium channels in HEK293 cells and in brain cells in vivo . After packaging these vectors into enhancer-AAVs and administering to mice, immunohistochemical analyses showed telencephalic GABAergic interneuron-specific and dose-dependent transgene biodistribution. These vectors conferred strong dose-dependent protection against postnatal mortality and seizures in two DS mouse models carrying independent loss-of-function alleles of Scn1a, at two independent research sites, supporting the robustness of this approach. No mortality or toxicity was observed in wild-type mice injected with single vectors expressing either the N-terminal or C-terminal halves of SCN1A , or the dual vector system targeting interneurons. In contrast, nonselective neuronal targeting of SCN1A conferred less rescue against mortality and presented substantial preweaning lethality. These findings demonstrate proof-of-concept that interneuron-specific AAV-mediated SCN1A gene replacement is sufficient for significant rescue in DS mouse models and suggest it could be an effective therapeutic approach for patients with DS.
Collapse
|
15
|
Ricobaraza A, Bunuales M, Gonzalez-Aparicio M, Fadila S, Rubinstein M, Vides-Urrestarazu I, Banderas J, Sola-Sevilla N, Sanchez-Carpintero R, Lanciego JL, Roda E, Honrubia A, Arnaiz P, Hernandez-Alcoceba R. Preferential expression of SCN1A in GABAergic neurons improves survival and epileptic phenotype in a mouse model of Dravet syndrome. J Mol Med (Berl) 2023; 101:1587-1601. [PMID: 37819378 PMCID: PMC10697872 DOI: 10.1007/s00109-023-02383-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
The SCN1A gene encodes the alpha subunit of a voltage-gated sodium channel (Nav1.1), which is essential for the function of inhibitory neurons in the brain. Mutations in this gene cause severe encephalopathies such as Dravet syndrome (DS). Upregulation of SCN1A expression by different approaches has demonstrated promising therapeutic effects in preclinical models of DS. Limiting the effect to inhibitory neurons may contribute to the restoration of brain homeostasis, increasing the safety and efficacy of the treatment. In this work, we have evaluated different approaches to obtain preferential expression of the full SCN1A cDNA (6 Kb) in GABAergic neurons, using high-capacity adenoviral vectors (HC-AdV). In order to favour infection of these cells, we considered ErbB4 as a surface target. Incorporation of the EGF-like domain from neuregulin 1 alpha (NRG1α) in the fiber of adenovirus capsid allowed preferential infection in cells lines expressing ErbB4. However, it had no impact on the infectivity of the vector in primary cultures or in vivo. For transcriptional control of transgene expression, we developed a regulatory sequence (DP3V) based on the Distal-less homolog enhancer (Dlx), the vesicular GABA transporter (VGAT) promoter, and a portion of the SCN1A gene. The hybrid DP3V promoter allowed preferential expression of transgenes in GABAergic neurons both in vitro and in vivo. A new HC-AdV expressing SCN1A under the control of this promoter showed improved survival and amelioration of the epileptic phenotype in a DS mouse model. These results increase the repertoire of gene therapy vectors for the treatment of DS and indicate a new avenue for the refinement of gene supplementation in this disease. KEY MESSAGES: Adenoviral vectors can deliver the SCN1A cDNA and are amenable for targeting. An adenoviral vector displaying an ErbB4 ligand in the capsid does not target GABAergic neurons. A hybrid promoter allows preferential expression of transgenes in GABAergic neurons. Preferential expression of SCN1A in GABAergic cells is therapeutic in a Dravet syndrome model.
Collapse
Affiliation(s)
- Ana Ricobaraza
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Maria Bunuales
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Manuela Gonzalez-Aparicio
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Saja Fadila
- Sackler Faculty of Medicine, Goldschleger Eye Research Institute, Tel Aviv University, Tel Aviv, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moran Rubinstein
- Sackler Faculty of Medicine, Goldschleger Eye Research Institute, Tel Aviv University, Tel Aviv, Israel
- 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
| | - Irene Vides-Urrestarazu
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Julliana Banderas
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Noemi Sola-Sevilla
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Rocio Sanchez-Carpintero
- University Clinic of Navarra, Dravet Syndrome Unit, Pediatric Neurology Unit, IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Jose Luis Lanciego
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Madrid, Spain
| | - Elvira Roda
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
| | - Adriana Honrubia
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
| | - Patricia Arnaiz
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
| | - Ruben Hernandez-Alcoceba
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain.
| |
Collapse
|
16
|
Whyte-Fagundes P, Vance A, Carroll A, Figueroa F, Manukyan C, Baraban SC. Testing of putative antiseizure drugs in a preclinical Dravet syndrome zebrafish model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.11.566723. [PMID: 38014342 PMCID: PMC10680609 DOI: 10.1101/2023.11.11.566723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Dravet syndrome (DS) is a severe genetic epilepsy primarily caused by de novo mutations in a voltage-activated sodium channel gene (SCN1A). Patients face life-threatening seizures that are largely resistant to available anti-seizure medications (ASM). Preclinical DS animal models are a valuable tool to identify candidate ASMs for these patients. Among these, scn1lab mutant zebrafish exhibiting spontaneous seizure-like activity are particularly amenable to large-scale drug screening. Prior screening in a scn1lab mutant zebrafish line generated using N-ethyl-Nnitrosourea (ENU) identified valproate, stiripentol, and fenfluramine e.g., Federal Drug Administration (FDA) approved drugs with clinical application in the DS population. Successful phenotypic screening in scn1lab mutant zebrafish consists of two stages: (i) a locomotion-based assay measuring high-velocity convulsive swim behavior and (ii) an electrophysiology-based assay, using in vivo local field potential (LFP) recordings, to quantify electrographic seizure-like events. Using this strategy more than 3000 drug candidates have been screened in scn1lab zebrafish mutants. Here, we curated a list of nine additional anti-seizure drug candidates recently identified in preclinical models: 1-EBIO, AA43279, chlorzoxazone, donepezil, lisuride, mifepristone, pargyline, soticlestat and vorinostat. First-stage locomotion-based assays in scn1lab mutant zebrafish identified only 1-EBIO, chlorzoxazone and lisuride. However, second-stage LFP recording assays did not show significant suppression of spontaneous electrographic seizure activity for any of the nine anti-seizure drug candidates. Surprisingly, soticlestat induced frank electrographic seizure-like discharges in wild-type control zebrafish. Taken together, our results failed to replicate clear anti-seizure efficacy for these drug candidates highlighting a necessity for strict scientific standards in preclinical identification of ASMs.
Collapse
Affiliation(s)
- P Whyte-Fagundes
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - A Vance
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - A Carroll
- Behavioral Neurosciences, Northeastern University, Boston, MA, USA
| | - F Figueroa
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - C Manukyan
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - S C Baraban
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
17
|
Manning A, Han V, Stephens A, Wang R, Bush N, Bard M, Ramirez JM, Kalume F. Elevated susceptibility to exogenous seizure triggers and impaired interneuron excitability in a mouse model of Leigh syndrome epilepsy. Neurobiol Dis 2023; 187:106288. [PMID: 37704057 PMCID: PMC10621616 DOI: 10.1016/j.nbd.2023.106288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/12/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023] Open
Abstract
Mutations in the NADH dehydrogenase (ubiquinone reductase) iron‑sulfur protein 4 (NDUFS4) gene, which encodes for a key structural subunit of the OXFOS complex I (CI), lead to the most common form of mitochondrial disease in children known as Leigh syndrome (LS). As in other mitochondrial diseases, epileptic seizures constitute one of the most significant clinical features of LS. These seizures are often very difficult to treat and are a sign of poor disease prognosis. Mice with whole-body Ndufs4 KO are a well-validated model of LS; they exhibit epilepsy and several other clinical features of LS. We have previously shown that mice with Ndufs4 KO in only GABAergic interneurons (Gad2-Ndufs4-KO) reproduce the severe epilepsy phenotype observed in the global KO mice. This observation indicated that these mice represent an excellent model of LS epilepsy isolated from other clinical manifestations of the disease. To further characterize this epilepsy phenotype, we investigated seizure susceptibility to selected exogenous seizure triggers in Gad2-Ndufs4-KO mice. Then, using electrophysiology, imaging, and immunohistochemistry, we studied the cellular, physiological, and neuroanatomical consequences of Ndufs4 KO in GABAergic interneurons. Homozygous KO of Ndufs4 in GABAergic interneurons leads to a prominent susceptibility to exogenous seizure triggers, impaired interneuron excitability and interneuron loss. Finally, we found that the hippocampus and cortex participate in the generation of seizure activity in Gad2-Ndufs4-KO mice. These findings further define the LS epilepsy phenotype and provide important insights into the cellular mechanisms underlying epilepsy in LS and other mitochondrial diseases.
Collapse
Affiliation(s)
- Arena Manning
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Victor Han
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Alexa Stephens
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Rose Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Nicholas Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Michelle Bard
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Jan M Ramirez
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America; Department of Neurological Surgery, University of Washington, Seattle, WA, United States of America
| | - Franck Kalume
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States of America; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America; Department of Neurological Surgery, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
18
|
Goff KM, Liebergall SR, Jiang E, Somarowthu A, Goldberg EM. VIP interneuron impairment promotes in vivo circuit dysfunction and autism-related behaviors in Dravet syndrome. Cell Rep 2023; 42:112628. [PMID: 37310860 PMCID: PMC10592464 DOI: 10.1016/j.celrep.2023.112628] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 04/01/2023] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
Dravet syndrome (DS) is a severe neurodevelopmental disorder caused by loss-of-function variants in SCN1A, which encodes the voltage-gated sodium channel subunit Nav1.1. We recently showed that neocortical vasoactive intestinal peptide interneurons (VIP-INs) express Nav1.1 and are hypoexcitable in DS (Scn1a+/-) mice. Here, we investigate VIP-IN function at the circuit and behavioral level by performing in vivo 2-photon calcium imaging in awake wild-type (WT) and Scn1a+/- mice. VIP-IN and pyramidal neuron activation during behavioral transition from quiet wakefulness to active running is diminished in Scn1a+/- mice, and optogenetic activation of VIP-INs restores pyramidal neuron activity to WT levels during locomotion. VIP-IN selective Scn1a deletion reproduces core autism-spectrum-disorder-related behaviors in addition to cellular- and circuit-level deficits in VIP-IN function, but without epilepsy, sudden death, or avoidance behaviors seen in the global model. Hence, VIP-INs are impaired in vivo, which may underlie non-seizure cognitive and behavioral comorbidities in DS.
Collapse
Affiliation(s)
- Kevin M Goff
- Medical Scientist Training Program (MSTP), The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sophie R Liebergall
- Medical Scientist Training Program (MSTP), The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Evan Jiang
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ala Somarowthu
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ethan M Goldberg
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Barbieri R, Nizzari M, Zanardi I, Pusch M, Gavazzo P. Voltage-Gated Sodium Channel Dysfunctions in Neurological Disorders. Life (Basel) 2023; 13:life13051191. [PMID: 37240836 DOI: 10.3390/life13051191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
The pore-forming subunits (α subunits) of voltage-gated sodium channels (VGSC) are encoded in humans by a family of nine highly conserved genes. Among them, SCN1A, SCN2A, SCN3A, and SCN8A are primarily expressed in the central nervous system. The encoded proteins Nav1.1, Nav1.2, Nav1.3, and Nav1.6, respectively, are important players in the initiation and propagation of action potentials and in turn of the neural network activity. In the context of neurological diseases, mutations in the genes encoding Nav1.1, 1.2, 1.3 and 1.6 are responsible for many forms of genetic epilepsy and for Nav1.1 also of hemiplegic migraine. Several pharmacological therapeutic approaches targeting these channels are used or are under study. Mutations of genes encoding VGSCs are also involved in autism and in different types of even severe intellectual disability (ID). It is conceivable that in these conditions their dysfunction could indirectly cause a certain level of neurodegenerative processes; however, so far, these mechanisms have not been deeply investigated. Conversely, VGSCs seem to have a modulatory role in the most common neurodegenerative diseases such as Alzheimer's, where SCN8A expression has been shown to be negatively correlated with disease severity.
Collapse
Affiliation(s)
| | - Mario Nizzari
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| | - Ilaria Zanardi
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| | - Michael Pusch
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| | - Paola Gavazzo
- Institute of Biophysics, Via de Marini 6, 16149 Genova, Italy
| |
Collapse
|
20
|
Stöber TM, Batulin D, Triesch J, Narayanan R, Jedlicka P. Degeneracy in epilepsy: multiple routes to hyperexcitable brain circuits and their repair. Commun Biol 2023; 6:479. [PMID: 37137938 PMCID: PMC10156698 DOI: 10.1038/s42003-023-04823-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 04/06/2023] [Indexed: 05/05/2023] Open
Abstract
Due to its complex and multifaceted nature, developing effective treatments for epilepsy is still a major challenge. To deal with this complexity we introduce the concept of degeneracy to the field of epilepsy research: the ability of disparate elements to cause an analogous function or malfunction. Here, we review examples of epilepsy-related degeneracy at multiple levels of brain organisation, ranging from the cellular to the network and systems level. Based on these insights, we outline new multiscale and population modelling approaches to disentangle the complex web of interactions underlying epilepsy and to design personalised multitarget therapies.
Collapse
Affiliation(s)
- Tristan Manfred Stöber
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, 44801, Bochum, Germany
- Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Goethe University, 60590, Frankfurt, Germany
| | - Danylo Batulin
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- CePTER - Center for Personalized Translational Epilepsy Research, Goethe University, 60590, Frankfurt, Germany
- Faculty of Computer Science and Mathematics, Goethe University, 60486, Frankfurt, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Peter Jedlicka
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus Liebig University Giessen, 35390, Giessen, Germany.
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
21
|
Medina E, Peterson S, Ford K, Singletary K, Peixoto L. Critical periods and Autism Spectrum Disorders, a role for sleep. Neurobiol Sleep Circadian Rhythms 2023; 14:100088. [PMID: 36632570 PMCID: PMC9826922 DOI: 10.1016/j.nbscr.2022.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Brain development relies on both experience and genetically defined programs. Time windows where certain brain circuits are particularly receptive to external stimuli, resulting in heightened plasticity, are referred to as "critical periods". Sleep is thought to be essential for normal brain development. Importantly, studies have shown that sleep enhances critical period plasticity and promotes experience-dependent synaptic pruning in the developing mammalian brain. Therefore, normal plasticity during critical periods depends on sleep. Problems falling and staying asleep occur at a higher rate in Autism Spectrum Disorder (ASD) relative to typical development. In this review, we explore the potential link between sleep, critical period plasticity, and ASD. First, we review the importance of critical period plasticity in typical development and the role of sleep in this process. Next, we summarize the evidence linking ASD with deficits in synaptic plasticity in rodent models of high-confidence ASD gene candidates. We then show that the high-confidence rodent models of ASD that show sleep deficits also display plasticity deficits. Given how important sleep is for critical period plasticity, it is essential to understand the connections between synaptic plasticity, sleep, and brain development in ASD. However, studies investigating sleep or plasticity during critical periods in ASD mouse models are lacking. Therefore, we highlight an urgent need to consider developmental trajectory in studies of sleep and plasticity in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elizabeth Medina
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Sarah Peterson
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Kaitlyn Ford
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Kristan Singletary
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Lucia Peixoto
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
22
|
Berecki G, Bryson A, Polster T, Petrou S. Biophysical characterization and modelling of SCN1A gain-of-function predicts interneuron hyperexcitability and a predisposition to network instability through homeostatic plasticity. Neurobiol Dis 2023; 179:106059. [PMID: 36868483 DOI: 10.1016/j.nbd.2023.106059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/11/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
SCN1A gain-of-function variants are associated with early onset developmental and epileptic encephalopathies (DEEs) that possess distinct clinical features compared to Dravet syndrome caused by SCN1A loss-of-function. However, it is unclear how SCN1A gain-of-function may predispose to cortical hyper-excitability and seizures. Here, we first report the clinical features of a patient carrying a de novo SCN1A variant (T162I) associated with neonatal-onset DEE, and then characterize the biophysical properties of T162I and three other SCN1A variants associated with neonatal-onset DEE (I236V) and early infantile DEE (P1345S, R1636Q). In voltage clamp experiments, three variants (T162I, P1345S and R1636Q) exhibited changes in activation and inactivation properties that enhanced window current, consistent with gain-of-function. Dynamic action potential clamp experiments utilising model neurons incorporating Nav1.1. channels supported a gain-of-function mechanism for all four variants. Here, the T162I, I236V, P1345S, and R1636Q variants exhibited higher peak firing rates relative to wild type and the T162I and R1636Q variants produced a hyperpolarized threshold and reduced neuronal rheobase. To explore the impact of these variants upon cortical excitability, we used a spiking network model containing an excitatory pyramidal cell (PC) and parvalbumin positive (PV) interneuron population. SCN1A gain-of-function was modelled by enhancing the excitability of PV interneurons and then incorporating three simple forms of homeostatic plasticity that restored pyramidal cell firing rates. We found that homeostatic plasticity mechanisms exerted differential impact upon network function, with changes to PV-to-PC and PC-to-PC synaptic strength predisposing to network instability. Overall, our findings support a role for SCN1A gain-of-function and inhibitory interneuron hyperexcitability in early onset DEE. We propose a mechanism through which homeostatic plasticity pathways can predispose to pathological excitatory activity and contribute to phenotypic variability in SCN1A disorders.
Collapse
Affiliation(s)
- Géza Berecki
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Alexander Bryson
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Department of Neurology, Austin Health, Heidelberg, VIC 3084, Australia
| | - Tilman Polster
- Krankenhaus Mara, Bethel Epilepsy Centre, Department of Epileptology, Medical School, Bielefeld University, Campus Bielefeld-Bethel, Bielefeld, Germany
| | - Steven Petrou
- Ion Channels and Disease Group, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Praxis Precision Medicines, Inc., Cambridge, MA 02142, USA; Department of the Florey Institute, University of Melbourne, Parkville, VIC 3050, Australia.
| |
Collapse
|
23
|
Salazar JJ, Satriano A, Matamoros JA, Fernández-Albarral JA, Salobrar-García E, López-Cuenca I, de Hoz R, Sánchez-Puebla L, Ramírez JM, Alonso C, Satta V, Hernández-Fisac I, Sagredo O, Ramírez AI. Retinal Tissue Shows Glial Changes in a Dravet Syndrome Knock-in Mouse Model. Int J Mol Sci 2023; 24:ijms24032727. [PMID: 36769051 PMCID: PMC9916888 DOI: 10.3390/ijms24032727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Dravet syndrome (DS) is an epileptic encephalopathy caused by mutations in the Scn1a gene encoding the α1 subunit of the Nav1.1 sodium channel, which is associated with recurrent and generalized seizures, even leading to death. In experimental models of DS, histological alterations have been found in the brain; however, the retina is a projection of the brain and there are no studies that analyze the possible histological changes that may occur in the disease. This study analyzes the retinal histological changes in glial cells (microglia and astrocytes), retinal ganglion cells (RGCs) and GABAergic amacrine cells in an experimental model of DS (Syn-Cre/Scn1aWT/A1783V) compared to a control group at postnatal day (PND) 25. Retinal whole-mounts were labeled with anti-GFAP, anti-Iba-1, anti-Brn3a and anti-GAD65/67. Signs of microglial and astroglial activation, and the number of Brn3a+ and GAD65+67+ cells were quantified. We found retinal activation of astroglial and microglial cells but not death of RGCs and GABAergic amacrine cells. These changes are similar to those found at the level of the hippocampus in the same experimental model in PND25, indicating a relationship between brain and retinal changes in DS. This suggests that the retina could serve as a possible biomarker in DS.
Collapse
Affiliation(s)
- Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Andrea Satriano
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - José A. Matamoros
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - José A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Lidia Sánchez-Puebla
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Medicina, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Cristina Alonso
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Valentina Satta
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Inés Hernández-Fisac
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Onintza Sagredo
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
- Correspondence: (O.S.); (A.I.R.)
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
- Correspondence: (O.S.); (A.I.R.)
| |
Collapse
|
24
|
Anderson LL, Bahceci DA, Hawkins NA, Everett-Morgan D, Banister SD, Kearney JA, Arnold JC. Heterozygous deletion of Gpr55 does not affect a hyperthermia-induced seizure, spontaneous seizures or survival in the Scn1a+/- mouse model of Dravet syndrome. PLoS One 2023; 18:e0280842. [PMID: 36701411 PMCID: PMC9879440 DOI: 10.1371/journal.pone.0280842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
A purified preparation of cannabidiol (CBD), a cannabis constituent, has been approved for the treatment of intractable childhood epilepsies such as Dravet syndrome. Extensive pharmacological characterization of CBD shows activity at numerous molecular targets but its anticonvulsant mechanism(s) of action is yet to be delineated. Many suggest that the anticonvulsant action of CBD is the result of G protein-coupled receptor 55 (GPR55) inhibition. Here we assessed whether Gpr55 contributes to the strain-dependent seizure phenotypes of the Scn1a+/- mouse model of Dravet syndrome. The Scn1a+/- mice on a 129S6/SvEvTac (129) genetic background have no overt phenotype, while those on a [129 x C57BL/6J] F1 background exhibit a severe phenotype that includes hyperthermia-induced seizures, spontaneous seizures and reduced survival. We observed greater Gpr55 transcript expression in the cortex and hippocampus of mice on the seizure-susceptible F1 background compared to those on the seizure-resistant 129 genetic background, suggesting that Gpr55 might be a genetic modifier of Scn1a+/- mice. We examined the effect of heterozygous genetic deletion of Gpr55 and pharmacological inhibition of GPR55 on the seizure phenotypes of F1.Scn1a+/- mice. Heterozygous Gpr55 deletion and inhibition of GPR55 with CID2921524 did not affect the temperature threshold of a thermally-induced seizure in F1.Scn1a+/- mice. Neither was there an effect of heterozygous Gpr55 deletion observed on spontaneous seizure frequency or survival of F1.Scn1a+/- mice. Our results suggest that GPR55 antagonism may not be a suitable anticonvulsant target for Dravet syndrome drug development programs, although future research is needed to provide more definitive conclusions.
Collapse
Affiliation(s)
- Lyndsey L. Anderson
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Discipline of Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| | - Dilara A. Bahceci
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Discipline of Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| | - Nicole A. Hawkins
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, United States of America
| | - Declan Everett-Morgan
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| | - Samuel D. Banister
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, School of Chemistry, The University of Sydney, Sydney, NSW, Australia
| | - Jennifer A. Kearney
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, United States of America
| | - Jonathon C. Arnold
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Discipline of Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
25
|
Reddy DS. Therapeutic and clinical foundations of cannabidiol therapy for difficult-to-treat seizures in children and adults with refractory epilepsies. Exp Neurol 2023; 359:114237. [PMID: 36206806 DOI: 10.1016/j.expneurol.2022.114237] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022]
Abstract
Novel and effective antiseizure medications are needed to treat refractory and rare forms of epilepsy. Cannabinoids, which are obtained from the cannabis plant, have a long history of medical use, including for neurologic conditions. In 2018, the US Food and Drug Administration approved the first phytocannabinoid, cannabidiol (CBD, Epidiolex), which is now indicated for severe seizures associated with three rare forms of developmental and epileptic encephalopathy: Dravet syndrome, Lennox-Gastaut syndrome, and tuberous sclerosis complex. Compelling evidence supports the efficacy of CBD in experimental models and patients with epilepsy. In randomized clinical trials, highly-purified CBD has demonstrated efficacy with an acceptable safety profile in children and adults with difficult-to-treat seizures. Although the underlying antiseizure mechanisms of CBD in humans have not yet been elucidated, the identification of novel antiseizure targets of CBD preclinically indicates multimodal mechanisms that include non-cannabinoid pathways. In addition to antiseizure effects, CBD possesses strong anti-inflammatory and neuroprotective activities, which might contribute to protective effects in epilepsy and other conditions. This article provides a succinct overview of therapeutic approaches and clinical foundations of CBD, emphasizing the clinical utility of CBD for the treatment of seizures associated with refractory and rare epilepsies. CBD has shown to be a safe and effective antiseizure medicine, demonstrating a broad spectrum of efficacy across multiple seizure types, including those associated with severe epilepsies with childhood onset. Despite such promise, there are many perils with CBD that hampers its widespread use, including limited understanding of pharmacodynamics, limited exposure-response relationship, limited information for seizure freedom with continued use, complex pharmacokinetics with drug interactions, risk of adverse effects, and lack of expert therapeutic guidelines. These scientific issues need to be resolved by further investigations, which would decide the unique role of CBD in the management of refractory epilepsy.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA; Texas A&M Health Institute of Pharmacology and Neurotherapeutics, School of Medicine, Texas A&M University, Bryan, TX, USA; Engineering Medicine, Intercollegiate School of Engineering Medicine, Texas A&M University, Houston, TX, USA; Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA; Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
26
|
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
|
27
|
Suvekbala V, Ramachandran H, Veluchamy A, Mascarenhas MAB, Ramprasath T, Nair MKC, Garikipati VNS, Gundamaraju R, Subbiah R. The Promising Epigenetic Regulators for Refractory Epilepsy: An Adventurous Road Ahead. Neuromolecular Med 2022:10.1007/s12017-022-08723-0. [DOI: 10.1007/s12017-022-08723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/13/2022] [Indexed: 10/14/2022]
|
28
|
Neuronal splicing regulator RBFOX3 mediates seizures via regulating Vamp1 expression preferentially in NPY-expressing GABAergic neurons. Proc Natl Acad Sci U S A 2022; 119:e2203632119. [PMID: 35951651 PMCID: PMC9388145 DOI: 10.1073/pnas.2203632119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epilepsy is a common neurological disorder, which has been linked to mutations or deletions of RNA binding protein, fox-1 homolog (Caenorhabditis elegans) 3 (RBFOX3)/NeuN, a neuronal splicing regulator. However, the mechanism of seizure mediation by RBFOX3 remains unknown. Here, we show that mice with deletion of Rbfox3 in gamma-aminobutyric acid (GABA) ergic neurons exhibit spontaneous seizures and high premature mortality due to increased presynaptic release, postsynaptic potential, neuronal excitability, and synaptic transmission in hippocampal dentate gyrus granule cells (DGGCs). Attenuating early excitatory gamma-aminobutyric acid (GABA) action by administering bumetanide, an inhibitor of early GABA depolarization, rescued premature mortality. Rbfox3 deletion reduced hippocampal expression of vesicle-associated membrane protein 1 (VAMP1), a GABAergic neuron-specific presynaptic protein. Postnatal restoration of VAMP1 rescued premature mortality and neuronal excitability in DGGCs. Furthermore, Rbfox3 deletion in GABAergic neurons showed fewer neuropeptide Y (NPY)-expressing GABAergic neurons. In addition, deletion of Rbfox3 in NPY-expressing GABAergic neurons lowered intrinsic excitability and increased seizure susceptibility. Our results establish RBFOX3 as a critical regulator and possible treatment path for epilepsy.
Collapse
|
29
|
Wei AD, Wakenight P, Zwingman TA, Bard AM, Sahai N, Willemsen MH, Schelhaas HJ, Stegmann APA, Verhoeven JS, de Man SA, Wessels MW, Kleefstra T, Shinde DN, Helbig KL, Basinger A, Wagner VF, Rodriguez-Buritica D, Bryant E, Millichap JJ, Millen KJ, Dobyns WB, Ramirez JM, Kalume FK. Human KCNQ5 de novo mutations underlie epilepsy and intellectual disability. J Neurophysiol 2022; 128:40-61. [PMID: 35583973 PMCID: PMC9236882 DOI: 10.1152/jn.00509.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We identified six novel de novo human KCNQ5 variants in children with motor/language delay, intellectual disability (ID), and/or epilepsy by whole exome sequencing. These variants, comprising two nonsense and four missense alterations, were functionally characterized by electrophysiology in HEK293/CHO cells, together with four previously reported KCNQ5 missense variants (Lehman A, Thouta S, Mancini GM, Naidu S, van Slegtenhorst M, McWalter K, Person R, Mwenifumbo J, Salvarinova R; CAUSES Study; EPGEN Study; Guella I, McKenzie MB, Datta A, Connolly MB, Kalkhoran SM, Poburko D, Friedman JM, Farrer MJ, Demos M, Desai S, Claydon T. Am J Hum Genet 101: 65-74, 2017). Surprisingly, all eight missense variants resulted in gain of function (GOF) due to hyperpolarized voltage dependence of activation or slowed deactivation kinetics, whereas the two nonsense variants were confirmed to be loss of function (LOF). One severe GOF allele (P369T) was tested and found to extend a dominant GOF effect to heteromeric KCNQ5/3 channels. Clinical presentations were associated with altered KCNQ5 channel gating: milder presentations with LOF or smaller GOF shifts in voltage dependence [change in voltage at half-maximal conduction (ΔV50) = ∼-15 mV] and severe presentations with larger GOF shifts in voltage dependence (ΔV50 = ∼-30 mV). To examine LOF pathogenicity, two Kcnq5 LOF mouse lines were created with CRISPR/Cas9. Both lines exhibited handling- and thermal-induced seizures and abnormal cortical EEGs consistent with epileptiform activity. Our study thus provides evidence for in vivo KCNQ5 LOF pathogenicity and strengthens the contribution of both LOF and GOF mutations to global pediatric neurological impairment, including ID/epilepsy.NEW & NOTEWORTHY Six novel de novo human KCNQ5 variants were identified from children with neurodevelopmental delay, intellectual disability, and/or epilepsy. Expression of these variants along with four previously reported KCNQ5 variants from a similar cohort revealed GOF potassium channels, negatively shifted in V50 of activation and/or delayed deactivation kinetics. GOF is extended to KCNQ5/3 heteromeric channels, making these the predominant channels affected in heterozygous de novo patients. Kcnq5 LOF mice exhibited seizures, consistent with in vivo pathogenicity.
Collapse
Affiliation(s)
- Aguan D Wei
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Paul Wakenight
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Theresa A Zwingman
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Angela M Bard
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Nikhil Sahai
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Marjolein H Willemsen
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Human Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Helenius J Schelhaas
- Department of Neurology, Academic Centre for Epileptology Kempenhaeghe, Heeze, The Netherlands
| | - Alexander P A Stegmann
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Judith S Verhoeven
- Department of Neurology, Academic Centre for Epileptology Kempenhaeghe, Heeze, The Netherlands
| | - Stella A de Man
- Department of Pediatrics, Amphia Hospital, Breda, The Netherlands.,Department of Human Genetics, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Marja W Wessels
- Department of Human Genetics, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Tjitske Kleefstra
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Katherine L Helbig
- Ambry Genetics, Aliso Viejo, California.,Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Alice Basinger
- Medical Genetics, Cook Children's Hospital, Fort Worth, Texas
| | - Victoria F Wagner
- Department of Pediatrics, University of Texas Health Science Center, Houston, Texas
| | | | - Emily Bryant
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - John J Millichap
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Epilepsy Center, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Kathleen J Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington.,Department of Neurology, University of Washington School of Medicine, Seattle, Washington
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Franck K Kalume
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington.,Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
30
|
Genetic therapeutic advancements for Dravet Syndrome. Epilepsy Behav 2022; 132:108741. [PMID: 35653814 DOI: 10.1016/j.yebeh.2022.108741] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 11/03/2022]
Abstract
Dravet Syndrome is a genetic epileptic syndrome characterized by severe and intractable seizures associated with cognitive, motor, and behavioral impairments. The disease is also linked with increased mortality mainly due to sudden unexpected death in epilepsy. Over 80% of cases are due to a de novo mutation in one allele of the SCN1A gene, which encodes the α-subunit of the voltage-gated ion channel NaV1.1. Dravet Syndrome is usually refractory to antiepileptic drugs, which only alleviate seizures to a small extent. Viral, non-viral genetic therapy, and gene editing tools are rapidly enhancing and providing new platforms for more effective, alternative medicinal treatments for Dravet syndrome. These strategies include gene supplementation, CRISPR-mediated transcriptional activation, and the use of antisense oligonucleotides. In this review, we summarize our current knowledge of novel genetic therapies that are currently under development for Dravet syndrome.
Collapse
|
31
|
Fouda MA, Ghovanloo MR, Ruben PC. Late sodium current: incomplete inactivation triggers seizures, myotonias, arrhythmias, and pain syndromes. J Physiol 2022; 600:2835-2851. [PMID: 35436004 DOI: 10.1113/jp282768] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
Acquired and inherited dysfunction in voltage-gated sodium channels underlies a wide range of diseases. "In addition to the defects in trafficking and expression, sodium channelopathies are also caused by dysfunction in one or several gating properties, for instance activation or inactivation. Disruption of the channel inactivation leads to the increased late sodium current, which is a common defect in seizure disorders, cardiac arrhythmias skeletal muscle myotonia and pain. An increase in late sodium current leads to repetitive action potential in neurons and skeletal muscles, and prolonged action potential duration in the heart. In this topical review, we compare the effects of late sodium current in brain, heart, skeletal muscle, and peripheral nerves. Abstract figure legend Shows cartoon illustration of general Nav channel transitions between (1) resting, (2) open, and (3) fast inactivated states. Disruption of the inactivation process exacerbates (4) late sodium currents. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | | | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
32
|
Martin P, Kümmerle A. Motor and behavioral phenotype of Dravet syndrome in adulthood. Epilepsy Behav 2022; 129:108601. [PMID: 35203016 DOI: 10.1016/j.yebeh.2022.108601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/24/2022]
Abstract
In a comparative cross-sectional study, 26 adult individuals with clinically typical, genetically confirmed Dravet syndrome (DS) and an equal number of individuals with early onset, problematic epilepsy, and intellectual disability (ID) of comparable severity were included. The aim of the study was to find out whether patients with DS could be clearly distinguished from the comparison group with regard to neurological and behavioral symptoms. Significant differences were found in that individuals with DS clearly more frequently exhibited a symptom cluster characterized by bradykinesia, hypomimia, hypophonia, (spastic) increased muscle tone, ataxia, sthenic perseveration, and a special interest in colors. To these symptoms must be added, according to the findings of previous examinations, mastication, camptocormia/antecollis on the one hand, and the tendency to visual hallucinations on the other hand, in order to define one neuropsychiatric phenotype of DS in adulthood. To these symptoms must be added, according to the findings of previous investigations, crouch gait with camptocormia/antecollis on the one hand, and the tendency to visual hallucinations on the other hand, in order to define one outlined neuropsychiatric phenotype of DS in adulthood.
Collapse
Affiliation(s)
- Peter Martin
- Séguin-Clinic for Persons with Severe Intellectual Disability, Epilepsy Centre Kork, Germany; Medical Faculty, University of Freiburg, Germany.
| | | |
Collapse
|
33
|
Kaneko K, Currin CB, Goff KM, Wengert ER, Somarowthu A, Vogels TP, Goldberg EM. Developmentally regulated impairment of parvalbumin interneuron synaptic transmission in an experimental model of Dravet syndrome. Cell Rep 2022; 38:110580. [PMID: 35354025 PMCID: PMC9003081 DOI: 10.1016/j.celrep.2022.110580] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 01/09/2022] [Accepted: 03/06/2022] [Indexed: 11/25/2022] Open
Abstract
Dravet syndrome is a neurodevelopmental disorder characterized by epilepsy, intellectual disability, and sudden death due to pathogenic variants in SCN1A with loss of function of the sodium channel subunit Nav1.1. Nav1.1-expressing parvalbumin GABAergic interneurons (PV-INs) from young Scn1a+/− mice show impaired action potential generation. An approach assessing PV-IN function in the same mice at two time points shows impaired spike generation in all Scn1a+/− mice at postnatal days (P) 16–21, whether deceased prior or surviving to P35, with normalization by P35 in surviving mice. However, PV-IN synaptic transmission is dysfunctional in young Scn1a+/− mice that did not survive and in Scn1a+/− mice ≥ P35. Modeling confirms that PV-IN axonal propagation is more sensitive to decreased sodium conductance than spike generation. These results demonstrate dynamic dysfunction in Dravet syndrome: combined abnormalities of PV-IN spike generation and propagation drives early disease severity, while ongoing dysfunction of synaptic transmission contributes to chronic pathology. Dravet syndrome is caused by variants in SCN1A with loss of function of Nav1.1 sodium channels. Kaneko et al. use the “mini-slice” to record at two developmental time points. Impaired spike generation of Nav1.1-expressing PV interneurons in Scn1a+/− mice is transient, while abnormalities of PV interneuron synaptic transmission persist.
Collapse
Affiliation(s)
- Keisuke Kaneko
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Christopher B Currin
- The Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, Austria
| | - Kevin M Goff
- Medical Scientist Training Program (MSTP), The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Eric R Wengert
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Ala Somarowthu
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Tim P Vogels
- The Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, Austria
| | - Ethan M Goldberg
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
34
|
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
|
35
|
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: 31] [Impact Index Per Article: 10.3] [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
|
36
|
Chronic partial TrkB activation reduces seizures and mortality in a mouse model of Dravet syndrome. Proc Natl Acad Sci U S A 2022; 119:2022726119. [PMID: 35165147 PMCID: PMC8851461 DOI: 10.1073/pnas.2022726119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 12/03/2022] Open
Abstract
Dravet syndrome (DS) is a severe childhood epileptic encephalopathy characterized by intractable seizures and comorbidities, including a high rate of premature mortality. DS is mainly caused by loss-of-function mutations of the Scn1a gene encoding sodium channel Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, causing DS phenotypes. Effective pharmacological therapy targeting defective PV interneurons is currently not available. This study demonstrated that early treatment with a partial TrkB receptor agonist, LM22A-4, increased Nav1.1 expression, improved PV interneuron function, and reduced seizure occurrence and mortality rate in DS mice, suggesting a potential therapy for DS. Dravet syndrome (DS) is one of the most severe childhood epilepsies, characterized by intractable seizures and comorbidities including cognitive and social dysfunction and high premature mortality. DS is mainly caused by loss-of-function mutations in the Scn1a gene encoding Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, contributing to DS phenotypes. Effective pharmacological therapy that targets defective PV interneurons is not available. The known role of brain-derived neurotrophic factor (BDNF) in the development and maintenance of interneurons, together with our previous results showing improved PV interneuronal function and antiepileptogenic effects of a TrkB receptor agonist in a posttraumatic epilepsy model, led to the hypothesis that early treatment with a TrkB receptor agonist might prevent or reduce seizure activity in DS mice. To test this hypothesis, we treated DS mice with LM22A-4 (LM), a partial agonist at the BDNF TrkB receptor, for 7 d starting at postnatal day 13 (P13), before the onset of spontaneous seizures. Results from immunohistochemistry, Western blot, whole-cell patch-clamp recording, and in vivo seizure monitoring showed that LM treatment increased the number of perisomatic PV interneuronal synapses around cortical pyramidal cells in layer V, upregulated Nav1.1 in PV neurons, increased inhibitory synaptic transmission, and decreased seizures and the mortality rate in DS mice. The results suggest that early treatment with a partial TrkB receptor agonist may be a promising therapeutic approach to enhance PV interneuron function and reduce epileptogenesis and premature death in DS.
Collapse
|
37
|
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
|
38
|
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
|
39
|
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] [MESH Headings] [Grants] [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
|
40
|
Huang CH, Hung PL, Fan PC, Lin KL, Hsu TR, Chou IJ, Ho CS, Chou IC, Lin WS, Lee IC, Fan HC, Chen SJ, Liang JS, Tu YF, Chang TM, Hu SC, Wong LC, Hung KL, Lee WT. Clinical spectrum and the comorbidities of Dravet syndrome in Taiwan and the possible molecular mechanisms. Sci Rep 2021; 11:20242. [PMID: 34642351 PMCID: PMC8511137 DOI: 10.1038/s41598-021-98517-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/03/2021] [Indexed: 11/09/2022] Open
Abstract
Dravet syndrome (DS) is an uncommon epilepsy syndrome that may negatively affect the patients and their caregivers. However, reliable and valid measures of its impact on caregivers and the characteristics of patients with DS in Taiwan are lacking. This study aimed to describe the characteristics of patients with DS and concerns of their caregivers and establish a baseline frequency of disease characteristics using a cross-sectional survey in Taiwan. We assessed the caregivers of patients with DS using an online anonymous questionnaire. The seizure frequency decreased with age, although lacking statistical significance. Vaccines show no influence on the condition of patients with DS. Our findings revealed the highest impact on the domains affecting the caregivers’ daily life, including additional household tasks, symptom observation, further medical plan, and financial issues. Caregivers also expressed concerns regarding the lack of independence/constant care, seizure control, speech/communication, and impacts on siblings because of long-term care of the patients in parents’ absence. Our findings highlight the significant effects of caring for a child with DS on the lives of their caregivers in Taiwan; these findings will help raise awareness regarding the needs of these families. Furthermore, we discussed the possible pathophysiological mechanisms of associated comorbidities.
Collapse
Affiliation(s)
- Chia-Hsuan Huang
- Division of Pediatric Neurology, Department of Pediatrics, National Taiwan University Hospital Yunlin Branch, Yunlin County, Taiwan
| | - Pi-Lien Hung
- Department of Pediatric Neurology, Chang Gung Memorial Hospital-Kaohsiung, Kaohsiung, Taiwan
| | - Pi-Chuan Fan
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuang-Lin Lin
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ting-Rong Hsu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - I-Jun Chou
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Che-Sheng Ho
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
| | - I-Ching Chou
- Division of Pediatrics Neurology, China Medical University Children's Hospital, Taichung, Taiwan
| | - Wei-Sheng Lin
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Inn-Chi Lee
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan
| | - Shyi-Jou Chen
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jao-Shwann Liang
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Yi-Fang Tu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tung-Ming Chang
- Department of Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Su-Ching Hu
- Department of Pediatrics, Cathay General Hospital, Taipei, Taiwan
| | - Lee-Chin Wong
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Pediatrics, Cathay General Hospital, Taipei, Taiwan
| | - Kun-Long Hung
- Department of Pediatrics, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Wang-Tso Lee
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Pediatric Neurology, National Taiwan University Children's Hospital, 8, Chung-Shan South Road, Taipei, 100, Taiwan.
| |
Collapse
|
41
|
Anderson LL, Heblinski M, Absalom NL, Hawkins NA, Bowen M, Benson MJ, Zhang F, Bahceci D, Doohan PT, Chebib M, McGregor IS, Kearney JA, Arnold JC. Cannabigerolic acid, a major biosynthetic precursor molecule in cannabis, exhibits divergent effects on seizures in mouse models of epilepsy. Br J Pharmacol 2021; 178:4826-4841. [PMID: 34384142 PMCID: PMC9292928 DOI: 10.1111/bph.15661] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 11/28/2022] Open
Abstract
Background and Purpose Cannabis has been used to treat epilepsy for millennia, with such use validated by regulatory approval of cannabidiol (CBD) for Dravet syndrome. Unregulated artisanal cannabis‐based products used to treat children with intractable epilepsies often contain relatively low doses of CBD but are enriched in other phytocannabinoids. This raises the possibility that other cannabis constituents might have anticonvulsant properties. Experimental Approach We used the Scn1a+/− mouse model of Dravet syndrome to investigate the cannabis plant for phytocannabinoids with anticonvulsant effects against hyperthermia‐induced seizures. The most promising, cannabigerolic acid (CBGA), was further examined against spontaneous seizures and survival in Scn1a+/− mice and in electroshock seizure models. Pharmacological effects of CBGA were surveyed across multiple drug targets. Key Results The initial screen identified three phytocannabinoids with novel anticonvulsant properties: CBGA, cannabidivarinic acid (CBDVA) and cannabigerovarinic acid (CBGVA). CBGA was most potent and potentiated the anticonvulsant effects of clobazam against hyperthermia‐induced and spontaneous seizures, and was anticonvulsant in the MES threshold test. However, CBGA was proconvulsant in the 6‐Hz threshold test and a high dose increased spontaneous seizure frequency in Scn1a+/− mice. CBGA was found to interact with numerous epilepsy‐relevant targets including GPR55, TRPV1 channels and GABAA receptors. Conclusion and Implications These results suggest that CBGA, CBDVA and CBGVA may contribute to the effects of cannabis‐based products in childhood epilepsy. Although these phytocannabinoids have anticonvulsant potential and could be lead compounds for drug development programmes, several liabilities would need to be overcome before CBD is superseded by another in this class.
Collapse
Affiliation(s)
- L L Anderson
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - M Heblinski
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - N L Absalom
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - N A Hawkins
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - M Bowen
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - M J Benson
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - F Zhang
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - D Bahceci
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - P T Doohan
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - M Chebib
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - I S McGregor
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Psychology, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - J A Kearney
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - J C Arnold
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Righes Marafiga J, Vendramin Pasquetti M, Calcagnotto ME. GABAergic interneurons in epilepsy: More than a simple change in inhibition. Epilepsy Behav 2021; 121:106935. [PMID: 32035792 DOI: 10.1016/j.yebeh.2020.106935] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/20/2022]
Abstract
The pathophysiology of epilepsy has been historically grounded on hyperexcitability attributed to the oversimplified imbalance between excitation (E) and inhibition (I) in the brain. The decreased inhibition is mostly attributed to deficits in gamma-aminobutyric acid-containing (GABAergic) interneurons, the main source of inhibition in the central nervous system. However, the cell diversity, the wide range of spatiotemporal connectivity, and the distinct effects of the neurotransmitter GABA especially during development, must be considered to critically revisit the concept of hyperexcitability caused by decreased inhibition as a key characteristic in the development of epilepsy. Here, we will discuss that behind this known mechanism, there is a heterogeneity of GABAergic interneurons with distinct functions and sources, which have specific roles in controlling the neural network activity within the recruited microcircuit and altered network during the epileptogenic process. This article is part of the Special Issue "NEWroscience 2018.
Collapse
Affiliation(s)
- Joseane Righes Marafiga
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Mayara Vendramin Pasquetti
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre 90046-900, RS, Brazil.
| |
Collapse
|
43
|
Abstract
Pathogenic variants in epilepsy genes result in a spectrum of clinical severity. One source of phenotypic heterogeneity is modifier genes that affect expressivity of a primary pathogenic variant. Mouse epilepsy models also display varying degrees of clinical severity on different genetic backgrounds. Mice with heterozygous deletion of Scn1a (Scn1a+/−) model Dravet syndrome, a severe epilepsy most often caused by SCN1A haploinsufficiency. Scn1a+/− mice recapitulate features of Dravet syndrome, including spontaneous seizures, sudden death, and cognitive/behavioral deficits. Scn1a+/− mice maintained on the 129S6/SvEvTac (129) strain have normal lifespan and no spontaneous seizures. In contrast, admixture with C57BL/6J (B6) results in epilepsy and premature lethality. We previously mapped Dravet Survival Modifier loci (Dsm1-Dsm5) responsible for strain-dependent differences in survival. Gabra2, encoding the GABAA α2 subunit, was nominated as a candidate modifier at Dsm1. Direct measurement of GABAA receptors found lower abundance of α2-containing receptors in hippocampal synapses of B6 mice relative to 129. We also identified a B6-specific single nucleotide deletion within Gabra2 that lowers mRNA and protein by nearly 50%. Repair of this deletion reestablished normal levels of Gabra2 expression. In this study, we used B6 mice with a repaired Gabra2 allele to evaluate Gabra2 as a genetic modifier of severity in Scn1a+/− mice. Gabra2 repair restored transcript and protein expression, increased abundance of α2-containing GABAA receptors in hippocampal synapses, and rescued epilepsy phenotypes of Scn1a+/− mice. These findings validate Gabra2 as a genetic modifier of Dravet syndrome, and support enhancing function of α2-containing GABAA receptors as treatment strategy for Dravet syndrome.
Collapse
|
44
|
Velíšková J, Marra C, Liu Y, Shekhar A, Park DS, Iatckova V, Xie Y, Fishman GI, Velíšek L, Goldfarb M. Early onset epilepsy and sudden unexpected death in epilepsy with cardiac arrhythmia in mice carrying the early infantile epileptic encephalopathy 47 gain-of-function FHF1(FGF12) missense mutation. Epilepsia 2021; 62:1546-1558. [PMID: 33982289 DOI: 10.1111/epi.16916] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Fibroblast growth factor homologous factors (FHFs) are brain and cardiac sodium channel-binding proteins that modulate channel density and inactivation gating. A recurrent de novo gain-of-function missense mutation in the FHF1(FGF12) gene (p.Arg52His) is associated with early infantile epileptic encephalopathy 47 (EIEE47; Online Mendelian Inheritance in Man database 617166). To determine whether the FHF1 missense mutation is sufficient to cause EIEE and to establish an animal model for EIEE47, we sought to engineer this mutation into mice. METHODS The Arg52His mutation was introduced into fertilized eggs by CRISPR (clustered regularly interspaced short palindromic repeats) editing to generate Fhf1R52H /F+ mice. Spontaneous epileptiform events in Fhf1R52H /+ mice were assessed by cortical electroencephalography (EEG) and video monitoring. Basal heart rhythm and seizure-induced arrhythmia were recorded by electrocardiography. Modulation of cardiac sodium channel inactivation by FHF1BR52H protein was assayed by voltage-clamp recordings of FHF-deficient mouse cardiomyocytes infected with adenoviruses expressing wild-type FHF1B or FHF1BR52H protein. RESULTS All Fhf1R52H /+ mice experienced seizure or seizurelike episodes with lethal ending between 12 and 26 days of age. EEG recordings in 19-20-day-old mice confirmed sudden unexpected death in epilepsy (SUDEP) as severe tonic seizures immediately preceding loss of brain activity and death. Within 2-53 s after lethal seizure onset, heart rate abruptly declined from 572 ± 16 bpm to 108 ± 15 bpm, suggesting a parasympathetic surge accompanying seizures that may have contributed to SUDEP. Although ectopic overexpression of FHF1BR52H in cardiomyocytes induced a 15-mV depolarizing shift in voltage of steady-state sodium channel inactivation and slowed the rate of channel inactivation, heart rhythm was normal in Fhf1R52H /+ mice prior to seizure. SIGNIFICANCE The Fhf1 missense mutation p.Arg52His induces epileptic encephalopathy with full penetrance in mice. Both Fhf1 (p.Arg52His) and Scn8a (p.Asn1768Asp) missense mutations enhance sodium channel Nav 1.6 currents and induce SUDEP with bradycardia in mice, suggesting an FHF1/Nav 1.6 functional axis underlying altered brain sodium channel gating in epileptic encephalopathy.
Collapse
Affiliation(s)
- Jana Velíšková
- Department of Cell Biology & Anatomy and Department of Neurology, New York Medical College, Valhalla, New York, USA.,Department of Obstetrics and Gynecology, New York Medical College, Valhalla, New York, USA.,Department of Neurology, New York Medical College, Valhalla, New York, USA
| | - Christopher Marra
- Department of Biological Sciences, Hunter College of City University of New York, New York, New York, USA.,Program in Biology, Graduate Center of City University of New York, New York, New York, USA
| | - Yue Liu
- Department of Biological Sciences, Hunter College of City University of New York, New York, New York, USA.,Program in Biology, Graduate Center of City University of New York, New York, New York, USA
| | - Akshay Shekhar
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York, USA
| | - David S Park
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York, USA
| | - Vasilisa Iatckova
- Department of Biological Sciences, Hunter College of City University of New York, New York, New York, USA
| | - Ying Xie
- Department of Biological Sciences, Hunter College of City University of New York, New York, New York, USA
| | - Glenn I Fishman
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, New York, USA
| | - Libor Velíšek
- Department of Cell Biology & Anatomy and Department of Neurology, New York Medical College, Valhalla, New York, USA.,Department of Neurology, New York Medical College, Valhalla, New York, USA.,Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University of New York, New York, New York, USA.,Program in Biology, Graduate Center of City University of New York, New York, New York, USA
| |
Collapse
|
45
|
Haigh JL, Adhikari A, Copping NA, Stradleigh T, Wade AA, Catta-Preta R, Su-Feher L, Zdilar I, Morse S, Fenton TA, Nguyen A, Quintero D, Agezew S, Sramek M, Kreun EJ, Carter J, Gompers A, Lambert JT, Canales CP, Pennacchio LA, Visel A, Dickel DE, Silverman JL, Nord AS. Deletion of a non-canonical regulatory sequence causes loss of Scn1a expression and epileptic phenotypes in mice. Genome Med 2021; 13:69. [PMID: 33910599 PMCID: PMC8080386 DOI: 10.1186/s13073-021-00884-0] [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: 12/23/2020] [Accepted: 04/06/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genes with multiple co-active promoters appear common in brain, yet little is known about functional requirements for these potentially redundant genomic regulatory elements. SCN1A, which encodes the NaV1.1 sodium channel alpha subunit, is one such gene with two co-active promoters. Mutations in SCN1A are associated with epilepsy, including Dravet syndrome (DS). The majority of DS patients harbor coding mutations causing SCN1A haploinsufficiency; however, putative causal non-coding promoter mutations have been identified. METHODS To determine the functional role of one of these potentially redundant Scn1a promoters, we focused on the non-coding Scn1a 1b regulatory region, previously described as a non-canonical alternative transcriptional start site. We generated a transgenic mouse line with deletion of the extended evolutionarily conserved 1b non-coding interval and characterized changes in gene and protein expression, and assessed seizure activity and alterations in behavior. RESULTS Mice harboring a deletion of the 1b non-coding interval exhibited surprisingly severe reductions of Scn1a and NaV1.1 expression throughout the brain. This was accompanied by electroencephalographic and thermal-evoked seizures, and behavioral deficits. CONCLUSIONS This work contributes to functional dissection of the regulatory wiring of a major epilepsy risk gene, SCN1A. We identified the 1b region as a critical disease-relevant regulatory element and provide evidence that non-canonical and seemingly redundant promoters can have essential function.
Collapse
Affiliation(s)
- Jessica L Haigh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Nycole A Copping
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Tyler Stradleigh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - A Ayanna Wade
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Rinaldo Catta-Preta
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Linda Su-Feher
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Iva Zdilar
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Sarah Morse
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Timothy A Fenton
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Anh Nguyen
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Diana Quintero
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Samrawit Agezew
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Michael Sramek
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Ellie J Kreun
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Jasmine Carter
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Andrea Gompers
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Jason T Lambert
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Cesar P Canales
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA, USA
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Diane E Dickel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA, USA
| | - Jill L Silverman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA.
- MIND Institute and Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA.
| | - Alex S Nord
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA.
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
46
|
Cannon SC. Epilepsy channelopathies go neddy: stabilizing NaV1.1 channels by neddylation. J Clin Invest 2021; 131:148370. [PMID: 33855971 DOI: 10.1172/jci148370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function mutations of SCN1A encoding the pore-forming α subunit of the NaV1.1 neuronal sodium channel cause a severe developmental epileptic encephalopathy, Dravet syndrome (DS). In this issue of the JCI, Chen, Luo, Gao, et al. describe a phenocopy for DS in mice deficient for posttranslational conjugation with neural precursor cell expressed, developmentally downregulated 8 (NEDD8) (neddylation), selectively engineered in inhibitory interneurons. Pursuing the possibility that this phenotype is also caused by loss of NaV1.1, Chen, Luo, Gao, and colleagues show that interneuron excitability and GABA release are impaired, NaV1.1 degradation rate is increased with a commensurate decrease of NaV1.1 protein, and NaV1.1 is a substrate for neddylation. These findings establish neddylation as a mechanism for stabilizing NaV1.1 subunits and suggest another pathomechanism for epileptic sodium channelopathy.
Collapse
|
47
|
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
|
48
|
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
|
49
|
Goff KM, Goldberg EM. A Role for Vasoactive Intestinal Peptide Interneurons in Neurodevelopmental Disorders. Dev Neurosci 2021; 43:168-180. [PMID: 33794534 PMCID: PMC8440337 DOI: 10.1159/000515264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/10/2021] [Indexed: 11/19/2022] Open
Abstract
GABAergic inhibitory interneurons of the cerebral cortex expressing vasoactive intestinal peptide (VIP-INs) are rapidly emerging as important regulators of network dynamics and normal circuit development. Several recent studies have also identified VIP-IN dysfunction in models of genetically determined neurodevelopmental disorders (NDDs). In this article, we review the known circuit functions of VIP-INs and how they may relate to accumulating evidence implicating VIP-INs in the mechanisms of prominent NDDs. We highlight recurring VIP-IN-mediated circuit motifs that are shared across cerebral cortical areas and how VIP-IN activity can shape sensory input, development, and behavior. Ultimately, we extract a set of themes that inform our understanding of how VIP-INs influence pathogenesis of NDDs. Using publicly available single-cell RNA sequencing data from the Allen Institute, we also identify several underexplored disease-associated genes that are highly expressed in VIP-INs. We survey these genes and their shared related disease phenotypes that may broadly implicate VIP-INs in autism spectrum disorder and intellectual disability rather than epileptic encephalopathy. Finally, we conclude with a discussion of the relevance of cell type-specific investigations and therapeutics in the age of genomic diagnosis and targeted therapeutics.
Collapse
Affiliation(s)
- Kevin M Goff
- Medical Scientist Training Program (MSTP), The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ethan M Goldberg
- Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- The Epilepsy NeuroGenetics Initiative, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Departments of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Departments of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
50
|
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: 59] [Impact Index Per Article: 14.8] [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
|