Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Cell Neurosci ; 126: 103875, 2023 09.
Article in English | MEDLINE | ID: mdl-37352898

ABSTRACT

Metabotropic glutamate receptor 6 (mGluR6) predominantly localizes to the postsynaptic sites of retinal ON-bipolar cells, at which it recognizes glutamate released from photoreceptors. The C-terminal domain (CTD) of mGluR6 contains a cluster of basic amino acids resembling motifs for endoplasmic reticulum (ER) retention. We herein investigated whether these basic residues are involved in regulating the subcellular localization of mGluR6 in 293T cells expressing mGluR6 CTD mutants using immunocytochemistry, immunoprecipitation, and flow cytometry. We showed that full-length mGluR6 localized to the ER and cell surface, whereas mGluR6 mutants with 15- and 20-amino acid deletions from the C terminus localized to the ER, but were deficient at the cell surface. We also demonstrated that the cell surface deficiency of mGluR6 mutants was rescued by introducing an alanine substitution at basic residues within the CTD. The surface-deficient mGluR6 mutant still did not localize to the cell surface and was retained in the ER when co-expressed with surface-expressible constructs, including full-length mGluR6, even though surface-deficient and surface-expressible constructs formed heteromeric complexes. The co-expression of the surface-deficient mGluR6 mutant reduced the surface levels of surface-expressible constructs. These results indicate that basic residues in the mGluR6 CTD served as ER retention signals. We suggest that exposed ER retention motifs in the aberrant assembly containing truncated or misfolded mGluR6 prevent these protein complexes from being transported to the cell surface.


Subject(s)
Receptors, Metabotropic Glutamate , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/metabolism , Glutamic Acid/metabolism , Endoplasmic Reticulum/metabolism
2.
J Neurochem ; 158(4): 837-848, 2021 08.
Article in English | MEDLINE | ID: mdl-33067823

ABSTRACT

Metabotropic glutamate receptor 6, mGluR6, interacts with scaffold proteins and Gßγ subunits via its intracellular C-terminal domain (CTD). The mGluR6 pathway is critically involved in the retinal processing of visual signals. We herein investigated whether the CTD (residues 840-871) was necessary for mGluR6 cell surface localization and G-protein coupling using mGluR6-CTD mutants with immunocytochemistry, surface biotinylation assays, and electrophysiological approaches. We used 293T cells and primary hippocampal neurons as model systems. We examined C-terminally truncated mGluR6 and showed that the removal of up to residue 858 did not affect surface localization or glutamate-induced G-protein-mediated responses, whereas a 15-amino acid deletion (Δ857-871) impaired these functions. However, a 21-amino acid deletion (Δ851-871) restored surface localization and glutamate-dependent responses, which were again attenuated when the entire CTD was removed. The sequence alignment of group III mGluRs showed conserved amino acids resembling an ER retention motif in the CTD. These results suggest that the intracellular CTD is required for the cell surface transportation and receptor function of mGluR6, whereas it may contain regulatory elements for intracellular trafficking and signaling.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Glutamate/metabolism , Amino Acids/metabolism , Animals , Biotinylation , Cell Line , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Gene Deletion , Glutamic Acid/pharmacology , Humans , Mutation/genetics , Rats , Receptors, Glutamate/genetics , Signal Transduction/genetics
3.
Neurobiol Dis ; 141: 104954, 2020 07.
Article in English | MEDLINE | ID: mdl-32445790

ABSTRACT

Dravet syndrome is a severe infantile-onset epileptic encephalopathy which begins with febrile seizures and is caused by heterozygous loss-of-function mutations of the voltage-gated sodium channel gene SCN1A. We designed a CRISPR-based gene therapy for Scn1a-haplodeficient mice using multiple guide RNAs (gRNAs) in the promoter regions together with the nuclease-deficient Cas9 fused to transcription activators (dCas9-VPR) to trigger the transcription of SCN1A or Scn1a in vitro. We tested the effect of this strategy in vivo using an adeno-associated virus (AAV) mediated system targeting inhibitory neurons and investigating febrile seizures and behavioral parameters. In both the human and mouse genes multiple guide RNAs (gRNAs) in the upstream, rather than downstream, promoter region showed high and synergistic activities to increase the transcription of SCN1A or Scn1a in cultured cells. Intravenous injections of AAV particles containing the optimal combination of 4 gRNAs into transgenic mice with Scn1a-haplodeficiency and inhibitory neuron-specific expression of dCas9-VPR at four weeks of age increased Nav1.1 expression in parvalbumin-positive GABAergic neurons, ameliorated their febrile seizures and improved their behavioral impairments. Although the usage of transgenic mice and rather modest improvements in seizures and abnormal behaviors hamper direct clinical application, our results indicate that the upregulation of Scn1a expression in the inhibitory neurons can significantly improve the phenotypes, even when applied after the juvenile stages. Our findings also suggest that the decrease in Nav1.1 is directly involved in the symptoms seen in adults with Dravet syndrome and open a way to improve this condition.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/physiopathology , Epilepsy/genetics , Epilepsy/physiopathology , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/physiology , Neurons/physiology , Animals , Behavior, Animal , CRISPR-Cas Systems , Disease Models, Animal , Epilepsies, Myoclonic/prevention & control , Epilepsy/prevention & control , Female , GABAergic Neurons/physiology , Genetic Therapy/methods , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
4.
Neurobiol Dis ; 112: 24-34, 2018 04.
Article in English | MEDLINE | ID: mdl-29337050

ABSTRACT

Loss of function mutations in the SCN1A gene, which encodes the voltage-gated sodium channel Nav1.1, have been described in the majority of Dravet syndrome patients presenting with epileptic seizures, hyperactivity, autistic traits, and cognitive decline. We previously reported predominant Nav1.1 expression in parvalbumin-expressing (PV+) inhibitory neurons in juvenile mouse brain and observed epileptic seizures in mice with selective deletion of Scn1a in PV+ cells mediated by PV-Cre transgene expression (Scn1afl/+/PV-Cre-TG). Here we investigate the behavior of Scn1afl/+/PV-Cre-TG mice using a comprehensive battery of behavioral tests. We observed that Scn1afl/+/PV-Cre-TG mice display hyperactive behavior, impaired social novelty recognition, and altered spatial memory. We also generated Scn1afl/+/SST-Cre-KI mice with a selective Scn1a deletion in somatostatin-expressing (SST+) inhibitory neurons using an SST-IRES-Cre knock-in driver line. We observed that Scn1afl/+/SST-Cre-KI mice display no spontaneous convulsive seizures and that Scn1afl/+/SST-Cre-KI mice have a lowered threshold temperature for hyperthermia-induced seizures, although their threshold values are much higher than those of Scn1afl/+/PV-Cre-TG mice. We finally show that Scn1afl/+/SST-Cre-KI mice exhibited no noticeable behavioral abnormalities. These observations suggest that impaired Nav1.1 function in PV+ interneurons is critically involved in the pathogenesis of hyperactivity, autistic traits, and cognitive decline, as well as epileptic seizures, in Dravet syndrome.


Subject(s)
Exploratory Behavior/physiology , Interpersonal Relations , NAV1.1 Voltage-Gated Sodium Channel/genetics , Parvalbumins/biosynthesis , Parvalbumins/genetics , Spatial Memory/physiology , Animals , Gene Deletion , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAV1.1 Voltage-Gated Sodium Channel/deficiency
5.
Biochem Biophys Res Commun ; 491(4): 1070-1076, 2017 09 30.
Article in English | MEDLINE | ID: mdl-28784306

ABSTRACT

Nav1.1 and Nav1.2 are the voltage-gated sodium channel pore-forming alpha I and II subunits, encoded by the genes SCN1A and SCN2A. Although mutations of both genes have similarly been described in patients with epilepsy, autism and/or intellectual disability, their expression sites in brain are largely distinct. Nav1.1 was shown to be expressed dominantly in parvalbumin (PV)-positive or somatostatin (SST)-positive inhibitory neurons and in a sparsely-distributed subpopulation of excitatory neurons. In contrast, Nav1.2 has been reported to be dominantly expressed in excitatory neurons. Here we show that Nav1.2 is also expressed in caudal ganglionic eminence (CGE)-derived inhibitory neurons, and expressions of Nav1.1 and Nav1.2 are mutually-exclusive in many of brain regions including neocortex, hippocampus, cerebellum, striatum and globus pallidus. In neocortex at postnatal day 15, in addition to the expression in excitatory neurons we show that Nav1.2 is expressed in reelin (RLN)-positive/SST-negative inhibitory neurons that are presumably single-bouquet cells because of their cortical layer I-limited distribution, and vasoactive intestinal peptide (VIP)-positive neurons that would be multipolar cell because of their layer I/II margin and layer VI distribution. Although Nav1.2 has previously been reported to be expressed in SST-positive cells, we here show that Nav1.2 is not expressed in either of PV-positive or SST-positive inhibitory neurons. PV-positive and SST-positive inhibitory neurons derive from medial ganglionic eminence (MGE) and innervate excitatory neurons, while VIP-positive and RLN-positive/SST-negative inhibitory neurons derive from CGE, innervate on inhibitory neurons and play disinhibitory roles in the neural network. Our results therefore indicate that, while Nav1.1 is expressed in MEG-derived inhibitory neurons, Nav1.2 is expressed in CGE-derived disinhibitory interneurons in addition to excitatory neurons. These findings should contribute to understanding of the pathology of neurodevelopmental diseases caused by SCN2A mutations.


Subject(s)
Interneurons/metabolism , NAV1.1 Voltage-Gated Sodium Channel/biosynthesis , NAV1.2 Voltage-Gated Sodium Channel/biosynthesis , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reelin Protein
6.
Hum Mol Genet ; 22(23): 4784-804, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23922229

ABSTRACT

Dravet syndrome is a severe epileptic encephalopathy mainly caused by heterozygous mutations in the SCN1A gene encoding a voltage-gated sodium channel Nav1.1. We previously reported dense localization of Nav1.1 in parvalbumin (PV)-positive inhibitory interneurons in mice and abnormal firing of those neurons in Nav1.1-deficient mice. In the present study, we investigated the physiologic consequence of selective Nav1.1 deletion in mouse global inhibitory neurons, forebrain excitatory neurons or PV cells, using vesicular GABA transporter (VGAT)-Cre, empty spiracles homolog 1 (Emx1)-Cre or PV-Cre recombinase drivers. We show that selective Nav1.1 deletion using VGAT-Cre causes epileptic seizures and premature death that are unexpectedly more severe than those observed in constitutive Nav1.1-deficient mice. Nav1.1 deletion using Emx1-Cre does not cause any noticeable abnormalities in mice; however, the severe lethality observed with VGAT-Cre-driven Nav1.1 deletion is rescued by additional Nav1.1 deletion using Emx1-Cre. In addition to predominant expression in PV interneurons, we detected Nav1.1 in subpopulations of excitatory neurons, including entorhino-hippocampal projection neurons, a subpopulation of neocortical layer V excitatory neurons, and thalamo-cortical projection neurons. We further show that even minimal selective Nav1.1 deletion, using PV-Cre, is sufficient to cause spontaneous epileptic seizures and ataxia in mice. Overall, our results indicate that functional impairment of PV inhibitory neurons with Nav1.1 haploinsufficiency contributes to the epileptic pathology of Dravet syndrome, and show for the first time that Nav1.1 haploinsufficiency in excitatory neurons has an ameliorating effect on the pathology.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/physiopathology , NAV1.1 Voltage-Gated Sodium Channel/physiology , Animals , Death, Sudden , Disease Models, Animal , Haploinsufficiency , Interneurons/metabolism , Interneurons/pathology , Mice , Mutation , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neurons/metabolism , Neurons/pathology , Parvalbumins/metabolism
7.
Neurobiol Dis ; 49: 29-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22986304

ABSTRACT

Dravet syndrome is an intractable epileptic encephalopathy characterized by early onset epileptic seizures followed by cognitive decline, hyperactivity, autistic behaviors and ataxia. Most Dravet syndrome patients possess heterozygous mutations of SCN1A gene encoding voltage-gated sodium channel αI subunit (Nav1.1). We have previously reported that mice heterozygous for a nonsense mutation in Scn1a developed early onset epileptic seizures. However, the learning ability and sociability of the mice remained to be investigated. In the present study, we subjected heterozygous Scn1a mice to a comprehensive behavioral test battery. We found that while heterozygous Scn1a mice had lowered spontaneous motor activity in home cage, they were hyperactive in novel environments. Moreover, the mice had low sociability and poor spatial learning ability that correspond to the autistic behaviors and cognitive decline seen in Dravet syndrome patients. These results suggest that Nav1.1 haploinsufficiency intrinsically contributes to not only epileptic seizures but also lowered sociability and learning impairment in heterozygous Scn1a mutant mice, as it should also be the case in patients with Dravet syndrome.


Subject(s)
Epilepsies, Myoclonic/psychology , Learning Disabilities/physiopathology , NAV1.1 Voltage-Gated Sodium Channel/deficiency , Social Behavior , Animals , Brain/physiopathology , Disease Models, Animal , Electrodes, Implanted , Electroencephalography , Grooming/physiology , Haploinsufficiency , Male , Maze Learning/physiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , NAV1.1 Voltage-Gated Sodium Channel/genetics , Reversal Learning/physiology , Rotarod Performance Test , Smell/physiology
8.
Elife ; 122023 05 23.
Article in English | MEDLINE | ID: mdl-37219072

ABSTRACT

Expressions of voltage-gated sodium channels Nav1.1 and Nav1.2, encoded by SCN1A and SCN2A genes, respectively, have been reported to be mutually exclusive in most brain regions. In juvenile and adult neocortex, Nav1.1 is predominantly expressed in inhibitory neurons while Nav1.2 is in excitatory neurons. Although a distinct subpopulation of layer V (L5) neocortical excitatory neurons were also reported to express Nav1.1, their nature has been uncharacterized. In hippocampus, Nav1.1 has been proposed to be expressed only in inhibitory neurons. By using newly generated transgenic mouse lines expressing Scn1a promoter-driven green fluorescent protein (GFP), here we confirm the mutually exclusive expressions of Nav1.1 and Nav1.2 and the absence of Nav1.1 in hippocampal excitatory neurons. We also show that Nav1.1 is expressed in inhibitory and a subpopulation of excitatory neurons not only in L5 but all layers of neocortex. By using neocortical excitatory projection neuron markers including FEZF2 for L5 pyramidal tract (PT) and TBR1 for layer VI (L6) cortico-thalamic (CT) projection neurons, we further show that most L5 PT neurons and a minor subpopulation of layer II/III (L2/3) cortico-cortical (CC) neurons express Nav1.1 while the majority of L6 CT, L5/6 cortico-striatal (CS), and L2/3 CC neurons express Nav1.2. These observations now contribute to the elucidation of pathological neural circuits for diseases such as epilepsies and neurodevelopmental disorders caused by SCN1A and SCN2A mutations.


Subject(s)
Neocortex , Mice , Animals , Mice, Transgenic , Neocortex/metabolism , Green Fluorescent Proteins/metabolism , Pyramidal Tracts , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neurons/physiology , Pyramidal Cells/metabolism
9.
Epilepsia ; 53(6): e111-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22525008

ABSTRACT

Generalized epilepsy with febrile seizures plus (GEFS+) and severe myoclonic epilepsy of infancy (SMEI) differ in their clinical severity and prognosis even though mutations of the Na(v) 1.1 sodium channel are responsible for both disorders. We compared the electrophysiologic properties of two mutant Na(v) 1.1 channels characterized by distinct amino acid substitutions at the same residue position: GEFS+ (A1685V) and SMEI (A1685D). Both the mutants showed complete loss of function when expressed alone. However, the function of A1685V can be partly rescued by the ß(1) subunit, consistently with a folding defect, whereas that of A1685D was not rescued. These electrophysiologic differences are consistent with the divergence in clinical severity between GEFS+ and SMEI.


Subject(s)
Epilepsies, Myoclonic/genetics , Membrane Potentials/genetics , Mutation, Missense/physiology , Nerve Tissue Proteins/physiology , Seizures, Febrile/genetics , Sodium Channels/physiology , Arginine/genetics , Aspartic Acid/genetics , Biophysics , Cell Line, Transformed , Electric Stimulation , Humans , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Sodium Channels/genetics , Transfection/methods , Valine/genetics
10.
Epilepsia ; 53(12): e200-3, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23148524

ABSTRACT

Dravet syndrome is a severe form of epileptic encephalopathy characterized by early onset epileptic seizures followed by ataxia and cognitive decline. Approximately 80% of patients with Dravet syndrome have been associated with heterozygous mutations in SCN1A gene encoding voltage-gated sodium channel (VGSC) α(I) subunit, whereas a homozygous mutation (p.Arg125Cys) of SCN1B gene encoding VGSC ß(I) subunit was recently described in a patient with Dravet syndrome. To further examine the involvement of homozygous SCN1B mutations in the etiology of Dravet syndrome, we performed mutational analyses on SCN1B in 286 patients with epileptic disorders, including 67 patients with Dravet syndrome who have been negative for SCN1A and SCN2A mutations. In the cohort, we found one additional homozygous mutation (p.Ile106Phe) in a patient with Dravet syndrome. The identified homozygous SCN1B mutations indicate that SCN1B is an etiologic candidate underlying Dravet syndrome.


Subject(s)
Epilepsies, Myoclonic/genetics , Mutation/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/genetics , DNA Mutational Analysis , Homozygote , Humans , Male , Microsatellite Repeats/genetics , Young Adult
11.
Epilepsia ; 53(7): 1140-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22578034

ABSTRACT

PURPOSE: We previously reported a mutant mouse carrying a severe myoclonic epilepsy in infancy (SMEI) mutation in Scn1a. In this study, we examined the susceptibility to hyperthermia-induced seizures of heterozygous Scn1a mutant mice (Scn1a(RX/+)) and wild-type (Scn1a(+/+) ) mice. Then we assessed the efficacy of stiripentol (STP) monotherapy versus STP and clobazam (CLB) combination therapy to prevent hyperthermia-induced seizures in Scn1a(RX/+) mice. METHODS: The seizure-inducing body temperatures in Scn1a(RX/+) mice and age-matched Scn1a(+/+) mice were compared in three age groups (1 month, 3-5 months, > 6 months). Then STP, CLB, or STP + CLB was administered intraperitoneally to Scn1a(RX/+) mice of two age groups (p1M, aged 1 month; p5M, aged 5-10 months). The efficacy of medications was assessed by comparing the seizure-inducing body temperature and the duration of seizures. KEY FINDINGS: The seizure-inducing body temperature was significantly lower in Scn1a(RX/+) than in Scn1a(+/+) mice for all age groups (p < 0.01). The seizure-inducing body temperature was significantly elevated after administration of STP in p1M (p < 0.05) but not in p5M (p > 0.05), and it was significantly elevated after administration of CLB in both age groups (p < 0.05). The seizure-inducing body temperature was significantly higher after administration of STP + CLB than after administration of CLB in p5M (p < 0.05). SIGNIFICANCE: Scn1a (RX/+) mice have increased susceptibility to hyperthermia-induced seizure in all age groups. STP monotherapy is effective in preventing hyperthermia-induced seizures in Scn1a(RX/+) mice aged 1 month, but not in those aged 5 months and older. When used in combination therapy with CLB, STP inhibits the metabolism of CLB and probably synergistically enhances the anticonvulsant effect in mice aged 1 month.


Subject(s)
Anticonvulsants/therapeutic use , Dioxolanes/therapeutic use , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/etiology , Hyperthermia, Induced/adverse effects , Seizures/drug therapy , Age Factors , Animals , Benzodiazepines/therapeutic use , Clobazam , Disease Models, Animal , Drug Therapy, Combination , Electroencephalography , Epilepsies, Myoclonic/genetics , Genetic Predisposition to Disease , Mice , Mice, Transgenic , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Seizures/etiology , Sodium Channels/genetics
12.
Hum Mutat ; 31(7): 820-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20506560

ABSTRACT

Mutations involving the voltage-gated sodium channel alpha(I) gene SCN1A are major genetic causes of childhood epileptic disorders, as typified by Dravet syndrome. Here we investigated the upstream regions of the SCN1A 5' noncoding exons and found two major regions with promoter activity. These two major promoters were simultaneously active in various brain regions and in most neurons. Using multiplex ligation-dependent probe amplification (MLPA) assays with probes for the 5' noncoding exons, their upstream regions, and all coding exons of SCN1A, we investigated 130 epileptic patients who did not show any SCN1A mutations by sequence analysis of all coding exons and exon-intron boundaries. Among 71 Dravet syndrome patients, we found two patients with heterozygous microdeletions removing the 5' noncoding exons and regions with promoter activity but not affecting the coding exons. We also identified four patients with deletions/duplication in the coding region. One patient with symptomatic focal epilepsy also showed a deletion in the coding region. This study provides the first case of microdeletion limited to the SCN1A 5' promoter region with the coding sequence preserved, and indicates the critical involvement of this upstream region in the molecular pathology of Dravet syndrome.


Subject(s)
Epilepsies, Myoclonic/genetics , Gene Deletion , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , Sodium Channels/genetics , 5' Flanking Region/genetics , Animals , Base Sequence , Blotting, Northern , Brain/metabolism , Cells, Cultured , Female , Gene Duplication , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Molecular Sequence Data , NAV1.1 Voltage-Gated Sodium Channel , Pedigree , Purkinje Cells/cytology , Purkinje Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Syndrome , Transcription Initiation Site
13.
Mol Autism ; 10: 15, 2019.
Article in English | MEDLINE | ID: mdl-30962870

ABSTRACT

Background: Mutations of the SCN2A gene encoding a voltage-gated sodium channel alpha-II subunit Nav1.2 are associated with neurological disorders such as epilepsy, autism spectrum disorders, intellectual disability, and schizophrenia. However, causal relationships and pathogenic mechanisms underlying these neurological defects, especially social and psychiatric features, remain to be elucidated. Methods: We investigated the behavior of mice with a conventional or conditional deletion of Scn2a in a comprehensive test battery including open field, elevated plus maze, light-dark box, three chambers, social dominance tube, resident-intruder, ultrasonic vocalization, and fear conditioning tests. We further monitored the effects of the positive allosteric modulator of AMPA receptors CX516 on these model mice. Results: Conventional heterozygous Scn2a knockout mice (Scn2aKO/+) displayed novelty-induced exploratory hyperactivity and increased rearing. The increased vertical activity was reproduced by heterozygous inactivation of Scn2a in dorsal-telencephalic excitatory neurons but not in inhibitory neurons. Moreover, these phenotypes were rescued by treating Scn2aKO/+ mice with CX516. Additionally, Scn2aKO/+ mice displayed mild social behavior impairment, enhanced fear conditioning, and deficient fear extinction. Neuronal activity was intensified in the medial prefrontal cortex of Scn2aKO/+ mice, with an increase in the gamma band. Conclusions: Scn2aKO/+ mice exhibit a spectrum of phenotypes commonly observed in models of schizophrenia and autism spectrum disorder. Treatment with the CX516 ampakine, which ameliorates hyperactivity in these mice, could be a potential therapeutic strategy to rescue some of the disease phenotypes.


Subject(s)
Anxiety/genetics , Autism Spectrum Disorder/genetics , Memory , NAV1.2 Voltage-Gated Sodium Channel/genetics , Psychomotor Agitation/genetics , Social Behavior , Animals , Anxiety/drug therapy , Autism Spectrum Disorder/drug therapy , Dioxoles/therapeutic use , Gamma Rhythm , Haploinsufficiency , Male , Membrane Transport Modulators/therapeutic use , Mice , Mice, Inbred C57BL , Phenotype , Piperidines/therapeutic use , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Psychomotor Agitation/drug therapy
14.
Nat Commun ; 10(1): 1917, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015467

ABSTRACT

STXBP1 and SCN2A gene mutations are observed in patients with epilepsies, although the circuit basis remains elusive. Here, we show that mice with haplodeficiency for these genes exhibit absence seizures with spike-and-wave discharges (SWDs) initiated by reduced cortical excitatory transmission into the striatum. Mice deficient for Stxbp1 or Scn2a in cortico-striatal but not cortico-thalamic neurons reproduce SWDs. In Stxbp1 haplodeficient mice, there is a reduction in excitatory transmission from the neocortex to striatal fast-spiking interneurons (FSIs). FSI activity transiently decreases at SWD onset, and pharmacological potentiation of AMPA receptors in the striatum but not in the thalamus suppresses SWDs. Furthermore, in wild-type mice, pharmacological inhibition of cortico-striatal FSI excitatory transmission triggers absence and convulsive seizures in a dose-dependent manner. These findings suggest that impaired cortico-striatal excitatory transmission is a plausible mechanism that triggers epilepsy in Stxbp1 and Scn2a haplodeficient mice.


Subject(s)
Corpus Striatum/metabolism , Munc18 Proteins/genetics , NAV1.2 Voltage-Gated Sodium Channel/genetics , Neocortex/metabolism , Seizures/genetics , Synaptic Transmission , Action Potentials/drug effects , Animals , Anticonvulsants/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dioxoles/pharmacology , Electroencephalography , Epilepsy, Absence/drug therapy , Epilepsy, Absence/genetics , Epilepsy, Absence/metabolism , Epilepsy, Absence/physiopathology , Ethosuximide/pharmacology , Gene Expression Regulation , Haploinsufficiency , Interneurons/drug effects , Interneurons/metabolism , Interneurons/pathology , Mice , Mice, Knockout , Munc18 Proteins/deficiency , NAV1.2 Voltage-Gated Sodium Channel/deficiency , Neocortex/drug effects , Neocortex/pathology , Neural Pathways/drug effects , Neural Pathways/metabolism , Piperidines/pharmacology , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Seizures/metabolism , Seizures/physiopathology , Seizures/prevention & control , Signal Transduction , Thalamus/drug effects , Thalamus/metabolism
15.
J Neurosci ; 27(22): 5903-14, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17537961

ABSTRACT

Loss-of-function mutations in human SCN1A gene encoding Nav1.1 are associated with a severe epileptic disorder known as severe myoclonic epilepsy in infancy. Here, we generated and characterized a knock-in mouse line with a loss-of-function nonsense mutation in the Scn1a gene. Both homozygous and heterozygous knock-in mice developed epileptic seizures within the first postnatal month. Immunohistochemical analyses revealed that, in the developing neocortex, Nav1.1 was clustered predominantly at the axon initial segments of parvalbumin-positive (PV) interneurons. In heterozygous knock-in mice, trains of evoked action potentials in these fast-spiking, inhibitory cells exhibited pronounced spike amplitude decrement late in the burst. Our data indicate that Nav1.1 plays critical roles in the spike output from PV interneurons and, furthermore, that the specifically altered function of these inhibitory circuits may contribute to epileptic seizures in the mice.


Subject(s)
Axons/chemistry , Epilepsy/genetics , Interneurons/chemistry , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Inhibition , Parvalbumins/biosynthesis , Sodium Channels/genetics , Sodium Channels/metabolism , Action Potentials/genetics , Animals , Axons/metabolism , Cell Line , Epilepsy/metabolism , Humans , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Mutant Strains , NAV1.1 Voltage-Gated Sodium Channel , Nerve Net/chemistry , Nerve Net/metabolism , Nerve Tissue Proteins/physiology , Neural Inhibition/genetics , Sodium Channels/physiology
16.
Nat Neurosci ; 21(7): 996-1003, 2018 07.
Article in English | MEDLINE | ID: mdl-29867081

ABSTRACT

An accumulating body of experimental evidence has implicated hippocampal replay occurring within sharp wave ripples (SPW-Rs) as crucial for learning and memory in healthy subjects. This raises speculation that neurological disorders impairing memory disrupt either SPW-Rs or their underlying neuronal activity. We report that mice heterozygous for the gene Scn2a, a site of frequent de novo mutations in humans with intellectual disability, displayed impaired spatial memory. While we observed no changes during encoding, to either single place cells or cell assemblies, we identified abnormalities restricted to SPW-R episodes that manifest as decreased cell assembly reactivation strengths and truncated hippocampal replay sequences. Our results suggest that alterations to hippocampal replay content may underlie disease-associated memory deficits.


Subject(s)
Hippocampus/physiopathology , Memory Disorders/genetics , Memory, Short-Term/physiology , NAV1.2 Voltage-Gated Sodium Channel/genetics , Spatial Memory/physiology , Action Potentials/physiology , Animals , Behavior, Animal/physiology , Heterozygote , Male , Memory Disorders/physiopathology , Mice , Mice, Knockout , Neural Pathways/physiopathology , Neurons/physiology , Sleep/physiology
17.
Epilepsy Res ; 147: 9-14, 2018 11.
Article in English | MEDLINE | ID: mdl-30176532

ABSTRACT

OBJECTIVE: To understand cerebral brain dysfunction in patients with Dravet syndrome (DS), we conducted a [18F]fluorodeoxyglucose-positron emission tomography (FDG-PET) study in patients with DS whose SCN1A gene variant was confirmed. METHODS: FDG-PET was performed on eight patients with DS. A SCN1A mutation analysis revealed missense variants in four patients and truncation variants in four patients. The patients' ages at the time of the PET study were 2, 2, 2, 3, 6, 13, 20, and 29 years old, respectively. The patients' developmental/intelligence quotient at the time of the PET study were 62, 52, 64, 35, 30, 15, and <25, respectively. The mean standardized uptake value (SUV) was calculated in four segments (frontal, temporal, parietal, and occipital) for the semi-quantitative analysis of 18F-FDG uptake. This value represents the average of the regions of interest in each lobe and was divided by the average SUV of the cerebellar hemisphere of each patient and compared between the patients with DS and the diseased controls. RESULTS: Glucose uptake in patients with DS decreased significantly, particularly in those ≥6 years old. Importantly, a comparison between the younger and older patients with DS revealed that glucose uptake was normal in patients who were ≤3 years (2, 2, 2, and 3 years), whereas a profound reduction in glucose uptake in the fronto-temporo-parietal-occipital cortices was observed in patients ≥ 6 years (6, 13, 20, and 29 years). Magnetic resonance imaging revealed no detectable atrophic legions or other changes in the cerebral cortices of patients ≥ 6 years of age. SIGNIFICANCE: The present study showed a remarkable reduction in cerebral glucose metabolism in multiple lobes for the first time, which became obvious after the late infantile period. These findings may indicate a functional neuroimaging aspect of epileptic encephalopathy of DS or a feature of the SCN1A variant itself.


Subject(s)
Epilepsies, Myoclonic/diagnostic imaging , Epilepsies, Myoclonic/genetics , Fluorodeoxyglucose F18/pharmacokinetics , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Positron-Emission Tomography , Adolescent , Adult , Cerebellum/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Child , Child, Preschool , Female , Functional Laterality , Glucose/metabolism , Humans , Male , Tomography Scanners, X-Ray Computed , Young Adult
18.
Commun Biol ; 1: 96, 2018.
Article in English | MEDLINE | ID: mdl-30175250

ABSTRACT

Mutations in the SCN2A gene encoding a voltage-gated sodium channel Nav1.2 are associated with epilepsies, intellectual disability, and autism. SCN2A gain-of-function mutations cause early-onset severe epilepsies, while loss-of-function mutations cause autism with milder and/or later-onset epilepsies. Here we show that both heterozygous Scn2a-knockout and knock-in mice harboring a patient-derived nonsense mutation exhibit ethosuximide-sensitive absence-like seizures associated with spike-and-wave discharges at adult stages. Unexpectedly, identical seizures are reproduced and even more prominent in mice with heterozygous Scn2a deletion specifically in dorsal-telencephalic (e.g., neocortical and hippocampal) excitatory neurons, but are undetected in mice with selective Scn2a deletion in inhibitory neurons. In adult cerebral cortex of wild-type mice, most Nav1.2 is expressed in excitatory neurons with a steady increase and redistribution from proximal (i.e., axon initial segments) to distal axons. These results indicate a pivotal role of Nav1.2 haplodeficiency in excitatory neurons in epilepsies of patients with SCN2A loss-of-function mutations.

19.
Mol Cell Biol ; 24(2): 846-55, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701755

ABSTRACT

U small nuclear RNAs (snRNAs) and mRNAs are both transcribed by RNA polymerase II (Pol II), but the snRNAs have unusual TATA-less promoters and are neither spliced nor polyadenylated; instead, 3' processing is directed by a highly conserved 3' end formation signal that requires initiation from an snRNA promoter. Here we show that the C-terminal domain (CTD) of Pol II is required for efficient U2 snRNA transcription, as it is for mRNA transcription. However, CTD kinase inhibitors, such as 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) and 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H7), that block mRNA elongation do not affect U2 transcription, although 3' processing of the U2 primary transcript is impaired. We show further that U2 transcription is preferentially inhibited by low doses of UV irradiation or actinomycin D, which induce CTD kinase activity, and that UV inhibition can be rescued by treatment with DRB or H7. We propose that Pol II complexes transcribing snRNAs and mRNAs have distinct CTD phosphorylation patterns. mRNA promoters recruit factors including kinases that hyperphosphorylate the CTD, and the CTD in turn recruits proteins needed for mRNA splicing and polyadenylation. We predict that snRNA promoters recruit factors including a CTD kinase(s) whose snRNA-specific phosphorylation pattern recruits factors required for promoter-coupled 3' end formation.


Subject(s)
RNA Polymerase II/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Base Sequence , Cell Line , DNA/genetics , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Phosphorylation , Promoter Regions, Genetic , Protein Kinase Inhibitors , Protein Kinases/metabolism , Protein Structure, Tertiary , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/chemistry , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/radiation effects , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects , Ultraviolet Rays
20.
Epilepsy Res ; 75(1): 46-51, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17507202

ABSTRACT

We investigated the roles of mutations in voltage-gated sodium channel alpha 1 subunit gene (SCN1A) in epilepsies and psychiatric disorders. The SCN1A gene was screened for mutations in three unrelated Japanese families with generalized epilepsy with febrile seizure plus (GEFS+), febrile seizure with myoclonic seizures, or intractable childhood epilepsy with generalized tonic-clonic seizures (ICEGTC). In the family with GEFS+, one individual was affected with panic disorder and seizures, and another individual was diagnosed with Asperger syndrome and seizures. The novel mutation V1366I was found in all probands and patients with psychiatric disorders of the three families. These results suggest that SCN1A mutations may confer susceptibility to psychiatric disorders in addition to variable epileptic seizures. Unidentified modifiers may play critical roles in determining the ultimate phenotype of patients with sodium channel mutations.


Subject(s)
Epilepsy/genetics , Genetic Predisposition to Disease , Mutation , Nerve Tissue Proteins/genetics , Sodium Channels/genetics , Adult , Child , DNA Mutational Analysis , Epilepsy/classification , Epilepsy/complications , Female , Humans , Isoleucine/genetics , Male , NAV1.1 Voltage-Gated Sodium Channel , Pedigree , Phenotype , Valine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL